55
Downloaded from https://journals.lww.com/pccmjournal by BhDMf5ePHKav1zEoum1tQfN4a+kJLhEZgbsIHo4XMi0hCywCX1AWnYQp/IlQrHD3iopPs8eUYyoQV+SE/i4rymJGhZjEwuf7voSwPT7jF2Q= on 02/23/2020 Copyright © 2020 by the Society of Critical Care Medicine and the World Federation of Pediatric Intensive and Critical Care Societies. Unauthorized reproduction of this article is prohibited Online Special Article e52 www.pccmjournal.org February 2020 • Volume 21 • Number 2 These guidelines are simultaneously being published in Pediatric Critical Care Medicine (DOI: 10.1097/PCC.0000000000002198) and Intensive Care Medicine (DOI: DOI: 10.1007/s00134-019-05878-6). DOI: 10.1097/PCC.0000000000002198 Surviving Sepsis Campaign International Guidelines for the Management of Septic Shock and Sepsis-Associated Organ Dysfunction in Children Scott L. Weiss, MD, MSCE, FCCM (Co-Vice Chair) 1 ; Mark J. Peters, MD, PhD (Co-Vice Chair) 2 ; Waleed Alhazzani, MD, MSc, FRCPC (Methodology Chair) 3 ; Michael S. D. Agus, MD, FCCM, FAAP 4 ; Heidi R. Flori, MD, FAAP 5 ; David P. Inwald, MB, BChir, FRCPCH, FFICM, PhD 6 ; Simon Nadel, MBBS, MRCP, FRCP 6 ; Luregn J. Schlapbach, FCICM, FMH-ICU, FMH-Paeds, FMH-Neonatology 7 ; Robert C. Tasker, MB BS, MA, AM, MD, FRCPHC, FRCP 4 ; Andrew C. Argent, MB BCh, MMed, MD (Paediatrics) 8 ; Joe Brierley, MD, MA 9 ; Joseph Carcillo, MD 10 ; Enitan D. Carrol, MB ChB, MD, FRCPCH, DTMH 11 ; Christopher L. Carroll, MD, MS, FCCM, FAAP 12 ; Ira M. Cheifetz, MD, FCCM 13 ; Karen Choong, MB, BCh, FRCP(C) (methodologist) 3 ; Jeffry J. Cies, PharmD, MPH, BCPS-AQ ID, BCPPS, FCCP, FCCM, FPPAG 14 ; Andrea T. Cruz, MD, MPH, FAAP 15 ; Daniele De Luca, MD, PhD 16,17 ; Akash Deep, MB BS, MD, FRCPCH 18 ; Saul N. Faust, MA, MB BS, FRCPCH, PhD, FHEA 19 ; Claudio Flauzino De Oliveira, MD, PhD 20 ; Mark W. Hall, MD, FCCM, FAAP 21 ; Paul Ishimine, MD, FACEP, FAAP 22 ; Etienne Javouhey, MD, PhD 23 ; Koen F. M. Joosten, MD, PhD 24 ; Poonam Joshi, PhD 25 ; Oliver Karam, MD, PhD 26 ; Martin C. J. Kneyber, MD, PhD, FCCM 27 ; Joris Lemson, MD, PhD 28 ; Graeme MacLaren, MD, MSc, FCCM 29 ; Nilesh M. Mehta, MD 4 ; Morten Hylander Møller, MD, PhD 30 ; Christopher J. L. Newth, MD, ChB, FRCPC, FRACP 31 ; Trung C. Nguyen, MD, FAAP 15 ; Akira Nishisaki, MD, MSCE, FAAP 1 ; Mark E. Nunnally, MD, FCCM (methodologist) 32 ; Margaret M. Parker, MD, MCCM, FAAP 33 ; Raina M. Paul, MD, FAAP 34 ; Adrienne G. Randolph, MD, MS, FCCM, FAAP 4 ; Suchitra Ranjit, MD, FCCM 35 ; Lewis H. Romer, MD 36 ; Halden F. Scott, MD, MSCS, FAAP, FACEP 37 ; Lyvonne N. Tume, BS, MSN, PhD, RN 38 ; Judy T. Verger, RN, PhD, CPNP-AC, FCCM, FAAN 1,39 ; Eric A. Williams, MD, MS, MMM, FCCM, FAAP 15 ; Joshua Wolf, MBBS, PhD, FRACP 40 ; Hector R. Wong, MD 41 ; Jerry J. Zimmerman, MD, PhD, FCCM 42 ; Niranjan Kissoon, MB BS, MCCM, FRCP(C), FAAP, FACPE (Co-Chair) 43 ; Pierre Tissieres, MD, DSc (Co-Chair) 16,44 The following sponsoring organizations with formal liaison appointees en- dorse this guideline: American Academy of Pediatrics; American Association of Critical-Care Nurses; American College of Chest Physicians; American College of Emergency Physicians; American Thoracic Society; Australian and New Zealand Intensive Care Society; Canadian Critical Care Society; European Society of Neonatal and Pediatric Intensive Care; Pediatric Infec- tious Diseases Society; Scandinavian Society of Anaesthesiology and Inten- sive Care Medicine; Society of Infectious Diseases Pharmacists; UK Sepsis Trust; World Federation of Pediatric Intensive and Critical Care Societies. Copyright © 2020 by the Society of Critical Care Medicine, the European Society of Intensive Care Medicine, and the World Federation of Pediatric Intensive and Critical Care Societies

Surviving Sepsis Campaign International Guidelines for the ... · In 2001, the Surviving Sepsis Campaign (SSC) was formed by the Society of Critical Care Medicine (SCCM), European

  • Upload
    others

  • View
    2

  • Download
    0

Embed Size (px)

Citation preview

Page 1: Surviving Sepsis Campaign International Guidelines for the ... · In 2001, the Surviving Sepsis Campaign (SSC) was formed by the Society of Critical Care Medicine (SCCM), European

Dow

nloadedfrom

https://journals.lww.com

/pccmjournalby

BhDMf5ePH

Kav1zEoum1tQ

fN4a+kJLhEZgbsIH

o4XMi0hC

ywCX1AW

nYQp/IlQ

rHD3iopPs8eU

YyoQV+SE/i4rym

JGhZjEw

uf7voSwPT7jF2Q

=on

02/23/2020

Downloadedfromhttps://journals.lww.com/pccmjournalbyBhDMf5ePHKav1zEoum1tQfN4a+kJLhEZgbsIHo4XMi0hCywCX1AWnYQp/IlQrHD3iopPs8eUYyoQV+SE/i4rymJGhZjEwuf7voSwPT7jF2Q=on02/23/2020

Copyright © 2020 by the Society of Critical Care Medicine and the World Federation of Pediatric Intensive and Critical Care Societies.Unauthorized reproduction of this article is prohibited

Copyright © 2020 by the Society of Critical Care Medicine and the World Federation of Pediatric Intensive and Critical Care Societies.Unauthorized reproduction of this article is prohibited

Online Special Article

e52 www.pccmjournal.org February 2020 • Volume 21 • Number 2

These guidelines are simultaneously being published in Pediatric Critical Care Medicine (DOI: 10.1097/PCC.0000000000002198) and Intensive Care Medicine (DOI: DOI: 10.1007/s00134-019-05878-6).

DOI: 10.1097/PCC.0000000000002198

Surviving Sepsis Campaign International Guidelines for the Management of Septic Shock and Sepsis-Associated Organ Dysfunction in Children

Scott L. Weiss, MD, MSCE, FCCM (Co-Vice Chair)1; Mark J. Peters, MD, PhD (Co-Vice Chair)2;

Waleed Alhazzani, MD, MSc, FRCPC (Methodology Chair)3; Michael S. D. Agus, MD, FCCM, FAAP4;

Heidi R. Flori, MD, FAAP5; David P. Inwald, MB, BChir, FRCPCH, FFICM, PhD6;

Simon Nadel, MBBS, MRCP, FRCP6;

Luregn J. Schlapbach, FCICM, FMH-ICU, FMH-Paeds, FMH-Neonatology7;

Robert C. Tasker, MB BS, MA, AM, MD, FRCPHC, FRCP4;

Andrew C. Argent, MB BCh, MMed, MD (Paediatrics)8; Joe Brierley, MD, MA9;

Joseph Carcillo, MD10; Enitan D. Carrol, MB ChB, MD, FRCPCH, DTMH11;

Christopher L. Carroll, MD, MS, FCCM, FAAP12; Ira M. Cheifetz, MD, FCCM13;

Karen Choong, MB, BCh, FRCP(C) (methodologist)3;

Jeffry J. Cies, PharmD, MPH, BCPS-AQ ID, BCPPS, FCCP, FCCM, FPPAG14;

Andrea T. Cruz, MD, MPH, FAAP15; Daniele De Luca, MD, PhD16,17; Akash Deep, MB BS, MD, FRCPCH18;

Saul N. Faust, MA, MB BS, FRCPCH, PhD, FHEA19; Claudio Flauzino De Oliveira, MD, PhD20;

Mark W. Hall, MD, FCCM, FAAP21; Paul Ishimine, MD, FACEP, FAAP22; Etienne Javouhey, MD, PhD23;

Koen F. M. Joosten, MD, PhD24; Poonam Joshi, PhD25; Oliver Karam, MD, PhD26;

Martin C. J. Kneyber, MD, PhD, FCCM27; Joris Lemson, MD, PhD28; Graeme MacLaren, MD, MSc, FCCM29;

Nilesh M. Mehta, MD4; Morten Hylander Møller, MD, PhD30;

Christopher J. L. Newth, MD, ChB, FRCPC, FRACP31; Trung C. Nguyen, MD, FAAP15;

Akira Nishisaki, MD, MSCE, FAAP1; Mark E. Nunnally, MD, FCCM (methodologist)32;

Margaret M. Parker, MD, MCCM, FAAP33; Raina M. Paul, MD, FAAP34;

Adrienne G. Randolph, MD, MS, FCCM, FAAP4; Suchitra Ranjit, MD, FCCM35; Lewis H. Romer, MD36;

Halden F. Scott, MD, MSCS, FAAP, FACEP37; Lyvonne N. Tume, BS, MSN, PhD, RN38;

Judy T. Verger, RN, PhD, CPNP-AC, FCCM, FAAN1,39; Eric A. Williams, MD, MS, MMM, FCCM, FAAP15;

Joshua Wolf, MBBS, PhD, FRACP40; Hector R. Wong, MD41; Jerry J. Zimmerman, MD, PhD, FCCM42;

Niranjan Kissoon, MB BS, MCCM, FRCP(C), FAAP, FACPE (Co-Chair)43;

Pierre Tissieres, MD, DSc (Co-Chair)16,44

The following sponsoring organizations with formal liaison appointees en-dorse this guideline: American Academy of Pediatrics; American Association of Critical-Care Nurses; American College of Chest Physicians; American College of Emergency Physicians; American Thoracic Society; Australian and New Zealand Intensive Care Society; Canadian Critical Care Society; European Society of Neonatal and Pediatric Intensive Care; Pediatric Infec-tious Diseases Society; Scandinavian Society of Anaesthesiology and Inten-sive Care Medicine; Society of Infectious Diseases Pharmacists; UK Sepsis Trust; World Federation of Pediatric Intensive and Critical Care Societies.

Copyright © 2020 by the Society of Critical Care Medicine, the European Society of Intensive Care Medicine, and the World Federation of Pediatric Intensive and Critical Care Societies

Page 2: Surviving Sepsis Campaign International Guidelines for the ... · In 2001, the Surviving Sepsis Campaign (SSC) was formed by the Society of Critical Care Medicine (SCCM), European

Copyright © 2020 by the Society of Critical Care Medicine and the World Federation of Pediatric Intensive and Critical Care Societies.Unauthorized reproduction of this article is prohibited

Online Special Article

Pediatric Critical Care Medicine www.pccmjournal.org e53

1Children’s Hospital of Philadelphia, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA.

2Great Ormond Street Hospital for Children, London, United Kingdom.3Department of Medicine, Division of Critical Care, and Department of Health Research Methods and Impact, McMaster University, Hamilton, ON, Canada.

4Department of Pediatrics (to Dr. Agus), Department of Anesthesiology, Critical Care and Pain (to Drs. Mehta and Randolph), Boston Children’s Hospital and Harvard Medical School, Boston, MA.

5C.S. Mott Children’s Hospital, Ann Arbor, MI.6St. Mary’s Hospital, London, United Kingdom.7Paediatric Critical Care Research Group, The University of Queensland and Queensland Children’s Hospital, Brisbane, QLD, Australia.

8Red Cross War Memorial Children’s Hospital and University of Cape Town, Cape Town, South Africa.

9Great Ormond Street Hospital for Children, London, United Kingdom.10Children’s Hospital of Pittsburgh, Pittsburgh, PA.11University of Liverpool, Liverpool, United Kingdom.12Connecticut Children’s Medical Center, Hartford, CT.13Duke Children’s, Durham, NC.14St. Christopher’s Hospital for Children, Philadelphia, PA.15Texas Children’s Hospital, Houston, TX.16Paris South University Hospitals—Assistance Publique Hopitaux de

Paris, Paris, France.17Physiopathology and Therapeutic Innovation Unit—INSERM U999,

South Paris-Saclay University, Paris, France.18King’s College Hospital, London, United Kingdom.19University Hospital Southampton NHS Foundation Trust and University

of Southampton, Southampton, United Kingdom.20The Latin America Sepsis Institute, São Paulo, Brazil.21Nationwide Children’s Hospital, Columbus, OH.22Rady Children’s Hospital, San Diego, CA.23Centre Hospitalier Universitaire de Lyon, Lyon, France.24Erasmus University Medical Center, Rotterdam, The Netherlands.25All India Institute of Medical Sciences, New Delhi, India.26Children’s Hospital of Richmond at VCU, Richmond, VA.27Beatrix Children’s Hospital, Groningen, The Netherlands.28Radboud University Medical Centre, Nijmegen, The Netherlands.29National University Health System, Singapore, and Royal Children’s

Hospital, Melbourne, VIC, Australia.30Rigshospitalet Hospital, Copenhagen, Denmark.31Children’s Hospital of Los Angeles, Los Angeles, CA.32New York University Langone Medical Center, New York, NY.33Stony Brook University, Stony Brook, NY.34Advocate Children’s Hospital, Park Ridge, IL.35Apollo Hospitals, Chennai, India.36Johns Hopkins Children’s Center, Baltimore, MD.37Children’s Hospital Colorado, Aurora, CO.38University of the West of England, Bristol, United Kingdom.39College of Nursing, University of Iowa, Iowa City, IA.40St. Jude Children’s Research Hospital, Memphis, TN.41Cincinnati Children’s Hospital, Cincinnati, OH.42Seattle Children’s Hospital, Seattle, WA.43British Columbia Children’s Hospital, Vancouver, British Columbia,

Canada.44Institute of Integrative Biology of the Cell-CNRS, CEA, Univ Paris Sud,

Gif-sur-Yvette, France.

The Society of Critical Care Medicine guidelines are intended for ge-neral information only, are not medical advice, and do not replace pro-fessional advice, which should be sought for any medical condition. The

full disclaimer for guidelines can be accessed at https://www.sccm.org/Research/Guidelines/Guidelines.

Supplemental digital content is available for this article. Direct URL cita-tions appear in the printed text and are provided in the HTML and PDF versions of this article on the journal’s website (http://journals.lww.com/pccmjournal).

For information regarding this article, E-mail: [email protected]

Objectives: To develop evidence-based recommendations for clinicians caring for children (including infants, school-aged chil-dren, and adolescents) with septic shock and other sepsis-asso-ciated organ dysfunction.Design: A panel of 49 international experts, representing 12 in-ternational organizations, as well as three methodologists and three public members was convened. Panel members assembled at key international meetings (for those panel members attend-ing the conference), and a stand-alone meeting was held for all panel members in November 2018. A formal conflict-of-interest policy was developed at the onset of the process and enforced throughout. Teleconferences and electronic-based discussion among the chairs, co-chairs, methodologists, and group heads, as well as within subgroups, served as an integral part of the guide-line development process.Methods: The panel consisted of six subgroups: recognition and management of infection, hemodynamics and resuscitation, ven-tilation, endocrine and metabolic therapies, adjunctive therapies, and research priorities. We conducted a systematic review for each Population, Intervention, Control, and Outcomes question to identify the best available evidence, statistically summarized the evidence, and then assessed the quality of evidence using the Grading of Recommendations Assessment, Development, and Evaluation approach. We used the evidence-to-decision frame-work to formulate recommendations as strong or weak, or as a best practice statement. In addition, “in our practice” statements were included when evidence was inconclusive to issue a recom-mendation, but the panel felt that some guidance based on prac-tice patterns may be appropriate.Results: The panel provided 77 statements on the management and resuscitation of children with septic shock and other sepsis-associated organ dysfunction. Overall, six were strong recom-mendations, 52 were weak recommendations, and nine were best-practice statements. For 13 questions, no recommendations could be made; but, for 10 of these, “in our practice” statements were provided. In addition, 49 research priorities were identified.Conclusions: A large cohort of international experts was able to achieve consensus regarding many recommendations for the best care of children with sepsis, acknowledging that most aspects of care had relatively low quality of evidence resulting in the frequent issuance of weak recommendations. Despite this challenge, these recommendations regarding the management of children with septic shock and other sepsis-associated organ dysfunction pro-vide a foundation for consistent care to improve outcomes and in-form future research. (Pediatr Crit Care Med 2020; 21:e52–e106)Key Words: evidence-based medicine; Grading of Recommendations Assessment, Development, and Evaluation criteria; guidelines; infection; pediatrics; sepsis; septic shock; Surviving Sepsis Campaign

Page 3: Surviving Sepsis Campaign International Guidelines for the ... · In 2001, the Surviving Sepsis Campaign (SSC) was formed by the Society of Critical Care Medicine (SCCM), European

Copyright © 2020 by the Society of Critical Care Medicine and the World Federation of Pediatric Intensive and Critical Care Societies.Unauthorized reproduction of this article is prohibited

Copyright © 2020 by the Society of Critical Care Medicine and the World Federation of Pediatric Intensive and Critical Care Societies.Unauthorized reproduction of this article is prohibited

Weiss et al

e54 www.pccmjournal.org February 2020 • Volume 21 • Number 2

Sepsis is a leading cause of morbidity, mortality, and healthcare utilization for children worldwide. Globally, an estimated 22 cases of childhood sepsis per 100,000

person-years and 2,202 cases of neonatal sepsis per 100,000 live births occur, translating into 1.2 million cases of childhood sepsis per year (1). More than 4% of all hospitalized patients less than 18 years and ~8% of patients admitted to PICUs in high-income countries have sepsis (2–6). Mortality for chil-dren with sepsis ranges from 4% to as high as 50%, depending on illness severity, risk factors, and geographic location (2, 3, 7–9). The majority of children who die of sepsis suffer from refractory shock and/or multiple organ dysfunction syndrome, with many deaths occurring within the initial 48 to 72 hours of treatment (10–13). Early identification and appropriate resus-citation and management are therefore critical to optimizing outcomes for children with sepsis.

In 2001, the Surviving Sepsis Campaign (SSC) was formed by the Society of Critical Care Medicine (SCCM), European Society of Intensive Care Medicine (ESICM), and the International Sepsis Forum. A primary aim of the SSC was to develop evidenced-based guidelines and recommendations for the resuscitation and management of patients with sepsis. The initial guidelines were published in 2004 and have been reviewed and updated every four years thereafter. Following the 2016 edition, SCCM and ESICM reaffirmed their commit-ment to evidence-based guidelines for all patients by form-ing separate task forces dedicated to guidelines for adults and children.

The objective of the SCCM/ESICM “Surviving Sepsis Campaign International Guidelines for the Management of Septic Shock and Sepsis-associated Organ Dysfunction in Children” is to provide guidance for clinicians caring for chil-dren (including infants, school-aged children, and adolescents) with septic shock and other sepsis-associated organ dysfunc-tion. We sought to leverage the expertise of a clinical and methodology team to create comprehensive evidence-based recommendations for the recognition and management of children with septic shock or other sepsis-associated acute organ dysfunction. Recommendations from these guidelines are based on the best current evidence but cannot replace the clinician’s decision-making capability when presented with a patient’s unique set of clinical variables. Recommendations are intended to guide “best practice” rather than to establish a treatment algorithm or to define standard of care. These guidelines are appropriate for treating septic shock and other sepsis-associated organ dysfunction in a hospital, emergency, or acute care setting, although some may be applicable else-where. Although recommendations were developed without consideration to availability of resources, we acknowledge that variation within and across healthcare systems and ge-ographic regions will determine the practical application of these guidelines.

Although several recommendations for the care of children with sepsis and septic shock have been previously published (14–16), these new guidelines are not intended to update or iterate on these prior documents. Instead, it was the aim of

SCCM/ESICM “Surviving Sepsis Campaign” to provide an ev-idence-based approach to the management of septic shock and other sepsis-associated organ dysfunction in children using a comprehensive and transparent methodologic approach by a panel with geographic and professional diversity.

METHODOLOGY

DefinitionsIn 2005, the International Pediatric Sepsis Consensus Confer-ence published definitions and criteria for sepsis, severe sepsis, and septic shock in children based on prevailing views of adult sepsis at the time with modifications for physiology based on age and maturational considerations (17). In 2016, new adult definitions and criteria were published (Sepsis-3) with “sepsis” defined as life-threatening organ dysfunction caused by a dys-regulated host response to infection and “septic shock” the subset of sepsis with circulatory and cellular/metabolic dys-function associated with a higher risk of mortality (18). The term “severe sepsis” was replaced by this new definition of sepsis. Although application of Sepsis-3 to children has been attempted (19, 20), formal revisions to the 2005 pediatric sepsis definitions remain pending (21). Therefore, the majority of studies used to establish evidence for these guidelines referred to the 2005 nomenclature in which severe sepsis was defined as 1) greater than or equal to 2 age-based systemic inflammatory response syndrome (SIRS) criteria, 2) confirmed or suspected invasive infection, and 3) cardiovascular dysfunction, acute respiratory distress syndrome (ARDS), or greater than or equal to 2 noncardiovascular organ system dysfunctions; and septic shock was defined as the subset with cardiovascular dysfunc-tion, which included hypotension, treatment with a vasoactive medication, or impaired perfusion. However, studies that de-fined sepsis as severe infection leading to life-threatening organ dysfunction were included even if criteria used to define sepsis deviated from the 2005 consensus definitions.

For the purposes of these guidelines, we define septic shock in children as severe infection leading to cardiovascular dys-function (including hypotension, need for treatment with a vasoactive medication, or impaired perfusion) and “sepsis-associated organ dysfunction” in children as severe infection leading to cardiovascular and/or noncardiovascular organ dys-function. Because several methods to identify acute organ dys-function in children are currently available (17, 19, 20, 22, 23), we chose not to require a specific definition or scheme for this purpose.

Scope of PatientsThe panel intended these guidelines to apply to all patients from greater than or equal to 37 weeks gestation at birth to 18 years old with severe sepsis or septic shock as defined by the 2005 International Pediatric Sepsis Consensus Conference or inclusive of severe infection leading to life-threatening organ dysfunction. Practically, all infants, children, and adolescents with septic shock or other sepsis-associated acute organ dys-function are included in this scope. For simplicity, we will

Page 4: Surviving Sepsis Campaign International Guidelines for the ... · In 2001, the Surviving Sepsis Campaign (SSC) was formed by the Society of Critical Care Medicine (SCCM), European

Copyright © 2020 by the Society of Critical Care Medicine and the World Federation of Pediatric Intensive and Critical Care Societies.Unauthorized reproduction of this article is prohibited

Online Special Article

Pediatric Critical Care Medicine www.pccmjournal.org e55

henceforth use the term “children” to refer to infants, school-aged children, and adolescents in these guidelines.

All recommendations apply to children with septic shock and other sepsis-associated acute organ dysfunction unless spe-cific qualifications, such as the subset with immune compro-mise, are included in the recommendation. Even though these guidelines are not intended to address the management of in-fection with or without SIRS when there is not associated acute organ dysfunction, we recognize that sepsis exists as a spectrum and some children without known acute organ dysfunction may still benefit from similar therapies as those with known organ dysfunction. Finally, acknowledging that neonatal sepsis, especially in premature babies, may have distinct pathology, bi-ology, and therapeutic considerations, newborns less than 37 weeks gestation are excluded from the scope of these guidelines. The panel sought to include term neonates (0–28 d) born at greater than or equal to 37 weeks gestation within the scope of these guidelines because these infants may be recognized and resuscitated outside of a newborn nursery or neonatal ICU. However, because the panel did not specifically address studies of neonates with perinatal infection or conditions that can be associated with neonatal sepsis (e.g., persistent pulmonary hy-pertension of the newborn), these guidelines do not address all management considerations for neonatal sepsis.

Application of Guidelines by Local Resource AvailabilityThe intended target users of these guidelines are health profes-sionals caring for children with septic shock or other sepsis-associated organ dysfunction in a hospital, emergency, or other acute care setting. However, we acknowledge that many of the recommendations are likely to apply to the care of children with septic shock and other sepsis-associated organ dysfunc-tion across a broad array of settings with adaptation to specific environments and resource availability.

These guidelines were largely developed without consider-ation of healthcare resources (with some specific exceptions, e.g., fluid resuscitation), although we realize that medical care for children with septic shock and other sepsis-associated organ dysfunction is necessarily carried out within the confines of locally available resources. The panel supports that these guidelines should constitute a general scheme of “best prac-tice,” but that translation to treatment algorithms or bundles and standards of care will need to account for variation in the availability of local healthcare resources. The panel acknowl-edges as well the need for future research to test the adaptation of interventions to locally available resources.

Funding and SponsorshipAll funding for the development of these guidelines was pro-vided by SCCM and ESICM. In addition, sponsoring organiza-tions provided support for their members’ involvement.

Selection and Organization of Panel MembersThe selection of panel members was based on their expertise in specific aspects of pediatric sepsis. Co-chairs and co-vice chairs

were appointed by the SCCM and ESICM governing bodies; panel members were then recommended by the co-chairs and co-vice chairs. Each panel member was required to be a prac-ticing healthcare professional with a focus on the acute and/or emergent care of critically ill children with septic shock or other sepsis-associated acute organ dysfunction. Broad international and multiprofessional representation from critical and inten-sive care medicine, emergency medicine, anesthesiology, neo-natology, and infectious disease with inclusion of physicians, nurses, pharmacists, and advanced practice providers as part of the working group was ensured. Three members from the lay public were also included with a role to ensure that patient, family, and caregivers’ opinions were considered in prioritiz-ing outcomes and finalizing recommendations that the clini-cians proposed during the development process. Panelists were recruited from a wide number of countries and healthcare systems, including representation from resource-limited geo-graphic areas. A demographically diverse panel with regard to sex, race, and geography was assembled. Members were then allocated to specific groups based on their expertise.

The methodology team included trained methodologists from McMaster University in Canada (W.A., K.C.) and New York University in the United States (M.E.N.). The team in-cluded methodologists with a health research methodology degree (MSc or PhD) and/or advanced methodology train-ing, all of whom are also practicing intensivists. The method-ology team provided methodological guidance and leadership throughout the guideline development process.

Question Development and Outcome PrioritizationThe panel was divided into groups: 1) recognition and man-agement of infection, 2) hemodynamics and resuscitation, 3) ventilation, 4) endocrine and metabolic therapies, and 5) ad-junctive therapies. A sixth subgroup was added to review re-search priorities in pediatric sepsis.

The co-chairs, co-vice chairs, and group heads made ini-tial selections of the topics. We included topics addressed in the 2016 SSC adult guidelines that were relevant to children, as well as other key pediatric topics discussed in previously pub-lished guidelines (14–16). The PICO format, which describes the population (P), intervention (I), control (C), and out-comes (O), was used for all guideline questions. Group heads, panel members, and methodologists reviewed and selected PICO questions considered important to guide care for chil-dren with septic shock or other sepsis-associated organ dys-function. Panel members proposed additional PICO questions of high priority and clinical relevance. For practical reasons, we excluded several issues pertaining to general acute or crit-ical illness that were not specific for sepsis (e.g., head-of-bed positioning during invasive mechanical ventilation) and have been addressed in other guidelines (e.g., Pediatric Acute Lung Injury Consensus Conference [PALICC]) (24). However, top-ics with particular relevance to children with septic shock or other sepsis-associated acute organ dysfunction were included in this guideline, even if there was evaluation of similar or overlapping topics in previous publications. The final decision

Page 5: Surviving Sepsis Campaign International Guidelines for the ... · In 2001, the Surviving Sepsis Campaign (SSC) was formed by the Society of Critical Care Medicine (SCCM), European

Copyright © 2020 by the Society of Critical Care Medicine and the World Federation of Pediatric Intensive and Critical Care Societies.Unauthorized reproduction of this article is prohibited

Copyright © 2020 by the Society of Critical Care Medicine and the World Federation of Pediatric Intensive and Critical Care Societies.Unauthorized reproduction of this article is prohibited

Weiss et al

e56 www.pccmjournal.org February 2020 • Volume 21 • Number 2

regarding PICO question inclusion was reached by discussion and consensus among the guideline panel leaders with input from panel members and the methodology team in each group.

In adherence with the Grading of Recommendations Assessment, Development, and Evaluation (GRADE) approach, panel members compiled a list of potential out-comes for each PICO question. Subsequently, we electronically surveyed panel members and asked them to rate each outcome on a scale of 1 (not important) to 9 (critically important). We selected only outcomes that were critical (mean of 7 or more) for decision-making, taking a patient’s perspective. In addition, we presented all selected outcomes to public members to ask for their input and feedback. The final list of PICO questions is provided in Supplemental Table 1 (Supplemental Digital Content 1, http://links.lww.com/PCC/B138).

Search Strategy and Evidence SummationFor each PICO question, a professional medical librarian formulated the search strategy with input from the group heads, panel members, and methodologists. Searches utilized a combination of controlled vocabulary (e.g., “sepsis,” “bacte-rial infections,” “critical illness,” “intensive care units,” “pedi-atrics,” “NICU,” “PICU,” “emergency service”) and key words (e.g., “toxic shock,” “blood poisoning,” “acute infection,” “child”) in the core search. Additional controlled vocabulary and key words were incorporated to create separate strategies specific to the question posed. Research design filters (e.g., systematic reviews/meta-analyses, randomized controlled trials, observational studies) were also applied as appro-priate. Only English language studies were included. No date restrictions were imposed on the searches, but we removed animal-only and opinion pieces from the results. The med-ical librarian searched a minimum of two major databases (e.g., Cochrane Library, PubMed/MEDLINE, or Embase) to identify relevant systematic reviews, clinical trials, and ob-servational studies published through May 1, 2017. As this was the inaugural version of these guidelines for children, all publications up through May 1, 2017, were considered. Key studies published after the conclusion of the initial literature search on May 1, 2017, were incorporated into the evidence synthesis if identified by panel members as important and relevant even if they were not part of the initial literature re-view. We excluded articles published in abstract form, in a language other than English, and those focused solely on pre-clinical data. Panel members, with input from methodolo-gists, used the Cochrane risk of bias tool to assess the risk of bias of randomized trials (25) and Newcastle-Ottawa Scale to assess risk of bias of nonrandomized studies (26).

When applicable, the methodologists used meta-analytic techniques to generate pooled estimates across two or more studies. For meta-analysis of randomized clinical trials (RCTs), we used random-effects model and inverse variance method to pool estimates across relevant studies. We reported relative risks (RRs) and 95% CI for binary outcomes, and mean dif-ference (MD) and 95% CI for continuous outcomes. For ob-servational data, we conducted meta-analyses if all individual

studies provided adjusted estimates and included both an in-tervention and a control arm using a random-effects model and inverse variance method to pool adjusted odds ratio (OR) across relevant studies All analyses were conducted using RevMan software (Review Manager, Version 5.3; Copenhagen, Denmark).

Formulation of RecommendationsThe GRADE approach principles guided the assessment of quality of evidence from high to very low based on six domains: 1) risk of bias, 2) inconsistency, 3) indirectness, 4) imprecision, 5) publication bias, and 6) assessment of the balance between benefit and harm, patients’ values and preferences, cost and resources, and feasibility and acceptability of the intervention (27). Methodologists performed initial assessments of quality of evidence and incorporated feedback from panel members to generate final evidence profiles using GRADEpro GDT (28).

The panel initially considered only research focused on pe-diatric patients using a hierarchy of evidence (Table 1). Studies focusing on children with septic shock and other sepsis-asso-ciated organ dysfunction were prioritized, although studies in-clusive of more general pediatric populations (e.g., all PICU patients) were considered for some questions on a case-by-case basis. If there were no studies or insufficient data in children with sepsis or general pediatric illness, evidence from studies of adult patients was considered using an a priori framework to determine appropriateness of indirect evidence (Fig. 1). Evidence from adult studies was generally down-graded due to the indirectness of the evidence.

In a series of webinars, methodologists reviewed the rele-vant data for each PICO question with panel members to for-mulate initial recommendations. Each of the groups used the Evidence-to-Decision (EtD) framework to facilitate transition from evidence to the final recommendation. The EtD frame-work ensured that panel members took into consideration not only the quality of evidence and magnitude of effect but also balance between benefits and harms, patients’ values and pref-erences, resources, cost, acceptability, and feasibility (28).

We classified recommendations as strong or weak using the language “We recommend…” or “We suggest…,” respec-tively. We judged a strong recommendation in favor of an intervention to have desirable effects of adherence that will clearly outweigh the undesirable effects. We judged a weak recommendation in favor of an intervention to have desirable consequences of adherence that will probably outweigh the undesirable consequences, but confidence is diminished either because the quality of evidence was low or the benefits and risks were closely balanced. The implications of calling a rec-ommendation strong or weak are shown in Table 2. A strong recommendation does not necessarily imply a standard of care, and circumstances may exist in which a strong recom-mendation cannot or should not be followed for an individual patient. We permitted strong recommendations “for” an in-tervention based on low or very low quality of evidence when the intervention had the potential to improve survival and there was low risk for immediate harm. We permitted strong

Page 6: Surviving Sepsis Campaign International Guidelines for the ... · In 2001, the Surviving Sepsis Campaign (SSC) was formed by the Society of Critical Care Medicine (SCCM), European

Copyright © 2020 by the Society of Critical Care Medicine and the World Federation of Pediatric Intensive and Critical Care Societies.Unauthorized reproduction of this article is prohibited

Online Special Article

Pediatric Critical Care Medicine www.pccmjournal.org e57

recommendations “against” an intervention based on low or very low quality of evidence when there was uncertain benefit but very likely or certain harm, including high costs (29).

Best practice statements (BPSs) were developed as ungraded strong recommendations within strict conditions suggested by the GRADE Working Group (Table 3) (30). BPS were issued when the evidence could not be summarized or assessed using GRADE methodology, but the benefit or harm was deemed unequivocal. In addition, when evidence was insufficient to make a recommendation, but the panel felt that some guid-ance based on current practice patterns may be appropriate, we issued an “in our practice” statement. The “in our practice statements” were developed through a survey of panelists to as-certain their state of current practice. As such, “in our practice” statements are intended only to describe current variation in care and are not meant to be construed as recommendations.

As new data are continuously generated, the SSC is commit-ted to ensuring that these guidelines are updated or affirmed every 4 years or sooner if breaking and relevant evidence becomes available.

Voting ProcessPanel members convened to review evidence and discuss recom-mendations in-person and through web conferences. Following

the formulation of initial recommendations through discus-sion within subgroups, all panelists received links to evidence profiles and polls created using SurveyMonkey (Palo Alto, CA) to indicate agreement, disagreement, or abstention. Only panel members without relevant conflicts of interest could vote. Voters could provide feedback for consideration in revising statements. Panelists also deliberated during face-to-face meet-ings, during which subgroups presented their draft statements for discussion. Up to three rounds of voting were conducted throughout this process of deliberation in an attempt to achieve final consensus. Acceptance of a statement required votes from 75% of the panel members with an 80% agreement threshold.

A summary of all statements determined by the panel is shown in Appendix 1. Evidence summaries and evidence profiles that informed the recommendations are included in the supplemental tables and figures (Supplemental Digital Content 1, http://links.lww.com/PCC/B138). Links to specific tables and figures appear within the relevant text.

Conflict of Interest PolicyConflict-of-interest (COI) disclosures were sought through the SCCM from all panelists and support personnel prior to commencing activities, with updates annually and as needed. The process relied solely on personal disclosure, with clarifica-tions sought when necessary, and centered primarily around potential financial conflicts. The co-vice chairs reviewed all COI disclosures in accordance with SCCM’s standard oper-ating procedures, sought clarification when necessary, and worked with the co-chairs to recommend appropriate recusals. There was no industry input into or support of the guideline development process. No panelists received honoraria for any role in the guidelines process. Only librarians and a supporting project manager received compensation for their work.

Seven individuals were identified with potential COIs, but only three were deemed relevant to the final list of questions included in the scope of this guideline. These individuals were asked to abstain from voting on the final recommendations involving the potential COI. In addition, panel members were asked to voluntarily abstain from voting on final recommenda-tions if they had a potential academic COI (e.g., grant application that could benefit from wording of a particular recommenda-tion), although all panel members were welcome to participate in the group discussions leading up to the final recommendation to ensure that input was available from relevant experts.

SCREENING, DIAGNOSIS, AND SYSTEMATIC MANAGEMENT OF SEPSIS

1) In children who present as acutely unwell, we suggest implementing systematic screening for timely recognition of septic shock and other sepsis-associated organ dysfunc-tion (weak recommendation, very low quality of evidence).

Remarks: Systematic screening needs to be tailored to the type of patients, resources, and procedures within each insti-tution. Evaluation for the effectiveness and sustainability of screening should be incorporated as part of this process.

TABLE 1. Determination of the Quality of Evidence

Underlying methodology

High: Systematic reviews, RCTs

Moderate: Downgraded RCTs or upgraded observational studies

Low: Well-conducted prospective observational cohort studies

Very low: Downgraded observational cohort studies, case-control studies, case series or expert opinion or other evidence

Factors that may decrease the strength of evidence

Methodologic features of available RCTs suggesting high likelihood of bias

Inconsistency of results, including problems with subgroup analyses

Indirectness of evidence (differing population, intervention, control, outcomes, comparison)

Imprecision of results

High likelihood of reporting bias

Factors that may increase the strength of evidence

Large magnitude of effect (direct evidence, relative risk > 2 with no plausible confounders)

Very large magnitude of effect with relative risk > 5 and no threats to validity (by two levels)

Dose-response gradient

RCT = randomized clinical trial.

Page 7: Surviving Sepsis Campaign International Guidelines for the ... · In 2001, the Surviving Sepsis Campaign (SSC) was formed by the Society of Critical Care Medicine (SCCM), European

Copyright © 2020 by the Society of Critical Care Medicine and the World Federation of Pediatric Intensive and Critical Care Societies.Unauthorized reproduction of this article is prohibited

Copyright © 2020 by the Society of Critical Care Medicine and the World Federation of Pediatric Intensive and Critical Care Societies.Unauthorized reproduction of this article is prohibited

Weiss et al

e58 www.pccmjournal.org February 2020 • Volume 21 • Number 2

Rationale: Systematic screening for sepsis in children is driven by the premise that earlier recognition will lead to more timely initiation of therapy, which will translate to improved morbidity and/or mortality. Screening tools are designed to in-crease reliability of sepsis recognition and empower health-care professionals to seek rapid medical review. Rapid recognition of sepsis through standardized screening and procedures to guide management of patients identified as at-risk for sepsis should be an essential component of sepsis quality improvement (QI) programs. Although the optimal method or tool for screening is unclear, we suggest that screening tools be adapted to the type of patients, resources, and processes within each institution.

Several studies demonstrating that institutional sepsis QI efforts improve outcomes have successfully incorporated screening tools (31–37). Most reported sepsis screens were designed to prompt clinicians to prioritize review of patients that had triggered the screen, hence the ultimate decision to treat or not remains with the clinician. Although RCTs have evaluated the role of systematic screening algorithms to rec-ognize clinical deterioration in children more generally (38), high-quality trials on pediatric sepsis recognition are lack-ing (39), and data are not sufficient to suggest any particular screening tool, although several have been published (40–42) or shared online (http://www.survivingsepsis.org/Resources/Pages/Protocols-and-Checklists.aspx). Single-institution studies demonstrate that an electronic health record (EHR)-based screening tool can yield high sensitivity and, when coupled with sequential clinician assessment, improved speci-ficity (43). For facilities that use an EHR, a step-wise approach

combining EHR-triggered alerts followed by clinician assess-ment has the potential to shorten the time to sepsis recogni-tion (41). Notably, no study was found on systematic sepsis screening in low- and middle-income countries meeting the PICO criteria.

Institutions should monitor and evaluate their prac-tice following implementation of sepsis screening (44). Robust QI balancing measures that should be assessed in-clude clinician response, anchoring bias, increased and/or inappropriate antimicrobial prescriptions, fluid overload, increased PICU admissions and transfers to higher levels of care, and healthcare utilization costs (45). Application of a screening tool requires ongoing optimization of sen-sitivity and specificity, continuous improvement efforts to maintain provider education and familiarity with the tool, and continual data acquisition to monitor implementation and increase utilization (42). Finally, screening tools must work well with existing or planned other early warning and rapid response systems (46, 47) that may also have inherent limitations (38, 48).

2) We were unable to issue a recommendation about using blood lactate values to stratify children with suspected septic shock or other sepsis-associated organ dysfunction into low- versus high-risk of having septic shock or sepsis. However, in our practice, if lactate levels can be rapidly obtained, we often measure blood lactate in children when evaluating for septic shock and other sepsis-associated organ dysfunction.

Figure 1. Framework to determine the appropriateness of using indirect evidence from studies of children without sepsis or from adults.

Page 8: Surviving Sepsis Campaign International Guidelines for the ... · In 2001, the Surviving Sepsis Campaign (SSC) was formed by the Society of Critical Care Medicine (SCCM), European

Copyright © 2020 by the Society of Critical Care Medicine and the World Federation of Pediatric Intensive and Critical Care Societies.Unauthorized reproduction of this article is prohibited

Online Special Article

Pediatric Critical Care Medicine www.pccmjournal.org e59

Rationale: Blood lactate levels provide a valuable indirect marker of tissue hypoperfusion (49). Although increased lac-tate levels are not specific, they provide a quantifiable sur-rogate for tissue hypoxia and can be rapidly obtained by point-of-care tests available in many settings. In adults, blood lactate greater than 2 mmol/L is now included within the op-erational definition of septic shock as an indication of cel-lular/metabolic dysfunction, and measurement of lactate is included in the Hour-1 Sepsis Bundle, with recommendations to repeat lactate measurement if the initial value exceeds 2 mmol/L (18, 50, 51). In children, several observational stud-ies have demonstrated an association of elevated blood lac-tate levels with adverse outcomes in septic shock (11, 52–54). However, the optimal threshold to define “hyperlactatemia” remains unclear. In a PICU study, the mortality rate for chil-dren with hypotension requiring vasopressors with lactate greater than 2 mmol/L was 32.0% compared with 16.1% if lactate was less than or equal to 2 mmol/L (11). Other studies

have shown that lactate levels greater than 4 mmol/L are consistently associated with mortality (52). Although blood lactate may be affected by the conditions of the blood draw (e.g., use of a tourniquet), both venous and arterial lactate measurements obtained have been shown to be independ-ently associated with mortality in children (55). In one pro-spective study in children, normalization of lactate within 2–4 hours of presentation was associated with decreased risk of persistent organ dysfunction (adjusted RR, 0.47; 95% CI, 0.29–0.78) (56). However, no RCTs have tested whether ini-tial or serial measurement of blood lactate directly informs evaluation and/or management in children. Lactate levels should therefore be interpreted as part of a more compre-hensive assessment of clinical status and perfusion.

3) We recommend implementing a protocol/guideline for management of children with septic shock or other sepsis-associated organ dysfunction (BPS).

Rationale: Institutional protocols have been shown to im-prove the speed and reliability of care for children with septic shock or other sepsis-associated organ dysfunction. Studies reported improvements in mortality, length of stay (LOS), duration of organ dysfunction, and development of new or progressive multiple organ dysfunction syndrome (8, 32–34, 36, 57–61). Most of these studies have focused on timely de-livery of a “bundle of therapies” (e.g., blood culture, fluid bolus, and antibiotics). For example, an analysis of 1,179 chil-dren with sepsis across 54 hospitals in New York State found that completion of a sepsis bundle within 1 hour was asso-ciated with lower risk-adjusted OR of in-hospital mortality (0.59; 95% CI, 0.38–0.93; p = 0.02) (8). In a recent single-insti-tution study, bundle-compliant care in 1,380 children with septic shock was associated with a five-times lower mortality

TABLE 2. Implications of the Strength of Recommendation

Category StrengthQuality of

EvidenceImplications

to PatientsImplications to Clinicians

Implications to Policymakers

Strong recommendation

Strong Usually high or moderate

Most individuals in this situation would want the recommended course of action, and only a small proportion would not

Most individuals should receive the recommended course of action. Formal decision aids are not likely to be needed to help individuals make decisions consistent with their values and preferences

Can be adapted as policy in most situations, including for use as performance indicators

Weak recommendation

Weak Any The majority of individuals in this situation would want the suggested course of action, but many would not

Different choices are likely to be appropriate for different patients, and therapy should be tailored to the individual patient’s circumstances, such as patients’ or family’s values and preferences

Policies will likely be variable

Best practice statement

Strong Ungraded Same as strong recommendation

Same as strong recommendation

Same as strong recommendation

In our practice statement

Not a recommen-dation

NA NA NA NA

NA = not applicable.

TABLE 3. Criteria for Best Practice Statement

Criteria for Best Practice Statement

1) Is the statement clear and actionable?

2) Is the message necessary?

3) Is the net benefit (or harm) unequivocal?

4) Is the evidence difficult to collect and summarize?

5) Is the rationale explicit?

6) Is this better to be formally GRADEd?

Modified from Guyatt et al (30).

Page 9: Surviving Sepsis Campaign International Guidelines for the ... · In 2001, the Surviving Sepsis Campaign (SSC) was formed by the Society of Critical Care Medicine (SCCM), European

Copyright © 2020 by the Society of Critical Care Medicine and the World Federation of Pediatric Intensive and Critical Care Societies.Unauthorized reproduction of this article is prohibited

Copyright © 2020 by the Society of Critical Care Medicine and the World Federation of Pediatric Intensive and Critical Care Societies.Unauthorized reproduction of this article is prohibited

Weiss et al

e60 www.pccmjournal.org February 2020 • Volume 21 • Number 2

(OR, 0.20; 95% CI, 0.07–0.53) (33). In another study, imple-mentation of a sepsis protocol led to a substantial increase in the proportion of children who no longer had organ dys-function on day 2 after presentation (adjusted OR, 4.2; 95% CI, 1.7–10.4) (34). However, it should be noted that protocols studied to date have variable components, many studies do not report adherence to specific items within protocols, and only a few studies have attempted to adjust for initial illness severity or other patient factors, making it difficult to summarize stud-ies using the GRADE approach. Therefore, because available evidence shows a strong and consistent association that ad-herence to protocols reduces variability in care and improves outcomes, we recommend implementing a protocol/guideline for management of children with septic shock or other sepsis-associated organ dysfunction as a best practice.

4) We recommend obtaining blood cultures before initiating antimicrobial therapy in situations where this does not sub-stantially delay antimicrobial administration (BPS).

Rationale: Blood cultures remain the most commonly used method to identify bacteremia. Identification of a blood-borne pathogen can have significant clinical implications on the type and duration of antimicrobial therapy and is an important mechanism to recognize multidrug-resistant pathogens (62). Thus, whenever possible, blood cultures should be obtained prior to initiation of antimicrobial therapy in children with se-vere sepsis or septic shock. Although no studies have directly measured the effect of blood cultures alone on outcome in pe-diatric sepsis, several observational studies have demonstrated that a bundled approach to initial resuscitation that includes early blood cultures is associated with improved outcomes (8, 31, 33). If collection of the blood cultures is likely to delay ad-ministration of antimicrobial therapy to the patient, then ad-ministration of antimicrobials should take precedence, in view of the impact of delayed antimicrobial administration on pa-tient outcomes (63). However, because blood cultures may be the only source of information identifying bacterial antibiotic susceptibility, it is important to make all reasonable efforts to collect blood cultures before timely antimicrobial administra-tion. The collection of other biological specimens to identify pathogens from nonblood sites (e.g., urine, cerebrospinal fluid, tracheal aspirate, bronchoalveolar lavage, drainage from col-lections) should also happen as soon as possible, and depend-ing on the suspected site of infection, such specimens may have a higher yield of pathogen identification than blood cultures. Clinicians should also consider the epidemiology of pedi-atric infections in relation to age, sex, and host factors, such as comorbidities (64, 65). Specific patterns of pediatric blood-stream infections relating to age and comorbidities are well known, and approximatively one out of three bacteremia epi-sodes are associated with organ dysfunction in a recent large population-based study (65).

Limitations of standard blood cultures include the time needed to grow and then identify pathogens and their anti-biotic sensitivities, as well as the effect of previous therapy on diagnostic yield. New molecular technologies are becoming

available to facilitate earlier and faster microbiological diagno-ses. Such techniques may be able to identify a range of patho-gens well before blood cultures are positive (66), and may potentially identify pathogens even after the administration of antimicrobial therapy. However, new molecular diagnos-tics are currently relatively expensive, are not sufficient for all pathogens and antibiotic sensitivities, and are not universally available.

ANTIMICROBIAL THERAPY

5) In children with septic shock, we recommend starting antimicrobial therapy as soon as possible, within 1 hour of recognition (strong recommendation, very low quality of evidence).

6) In children with sepsis-associated organ dysfunction but without shock, we suggest starting antimicrobial therapy as soon as possible after appropriate evaluation, within 3 hours of recognition (weak recommendation, very low quality of evidence).

Rationale: Antimicrobials are the primary medical therapy that directly targets the underlying cause of sepsis, and there is strong biologic rationale for rapid delivery of antimicrobials in patients with sepsis (44). Many QI initiatives have shown improved pediatric sepsis outcomes with implementation of a bundle that includes rapid delivery of IV antimicrobials (8, 32–34, 36, 57–61). Two retrospective observational studies have also demonstrated an association of faster time to antimicrobial therapy with reduced mortality for children with sepsis. The first study was an analysis of 130 children with sepsis (mortality of 12%), including 103 (79%) with septic shock, in which the unadjusted OR for mortality among children with antimicro-bials delivered within versus after 60 minutes of sepsis recogni-tion was 0.60 (95% CI, 0.13–2.86) (63). The second study was an analysis of 1,179 children, including 69% with septic shock, where completion of a sepsis bundle within 1 hour of sepsis recognition was associated with decreased mortality (OR, 0.59; 95% CI, 0.38–0.93; p = 0.02); however, initiation of antimicro-bials alone by 1 hour of recognition was not associated with significant mortality reduction (OR, 0.78; 95% CI, 0.55–1.12; p = 0.18) (8). When the adjusted OR of these two studies were pooled, there was a possible reduction in mortality (OR, 0.77; 95% CI, 0.55–1.08) (Supplemental Table 2 and Supplemental Fig. 1, Supplemental Digital Content 1, http://links.lww.com/PCC/B138). Other secondary endpoints reported in the liter-ature have also been associated with shorter time to initiation of antimicrobial therapy, including reduced LOS, shorter du-ration of organ dysfunction, and reduced development of new or progressive multiple organ dysfunction syndrome (8, 32–34, 36, 57–61). Furthermore, indirect evidence from adult sepsis generally supports a benefit to starting antimicrobial therapy as soon as possible after recognition of septic shock (67–72). Thus, timely antimicrobial therapy—ideally administered as part of a more comprehensive bundle of initial care—should be the goal for children with septic shock.

Page 10: Surviving Sepsis Campaign International Guidelines for the ... · In 2001, the Surviving Sepsis Campaign (SSC) was formed by the Society of Critical Care Medicine (SCCM), European

Copyright © 2020 by the Society of Critical Care Medicine and the World Federation of Pediatric Intensive and Critical Care Societies.Unauthorized reproduction of this article is prohibited

Online Special Article

Pediatric Critical Care Medicine www.pccmjournal.org e61

The definition of “timely” in this context represents an area of controversy relating to challenges in the accurate recogni-tion of patients with sepsis and septic shock and the need to consider balancing QI metrics such as unnecessary antimicro-bial usage (44, 73, 74). One pediatric study (63) indicated a dose-response gradient such that the longer time to antimicro-bial therapy, the higher the mortality. Yet the mortality increase reached significance only when antimicrobials were adminis-tered greater than 3 hours in comparison to less than 3 hours, whereas the mortality of patients receiving antimicrobials within less than 1 hour was not different from those receiving antimicrobials within less than 3 hours in that relatively small study. The second, larger pediatric study demonstrated a sig-nificant decrease in mortality if antimicrobials were adminis-tered within 1 hour, but only in the context of a bundle that included a blood culture and fluid bolus (8). Thus, available pediatric studies do not provide a clear time cutoff after which the risk of mortality or other adverse outcomes increases, but rather support that there is likely to be an incremental risk for harm as time to antimicrobial initiation increases, in par-ticular beyond 3 hours. Notably, the benefit of antimicrobial therapy within 1 hour of recognition has been most prominent in cohorts with a predominance of septic shock (as compared with sepsis without shock) patients (8, 63).

Based on limited pediatric evidence and indirect evidence from adult studies, the panel supported that, in children “with septic shock,” antimicrobial therapy should be initiated as soon as possible and ideally within 1 hour of recognition. Suspicion of septic shock can usually be guided by clinical findings rap-idly ascertained through history and physical examination. Although our recommendation to ideally administer antimi-crobial administration within 1 hour of recognition of septic shock establishes a tangible goal that emphasizes the impor-tance of early antimicrobial therapy and assists clinicians in prioritizing bedside care, this cut-point should not be miscon-strued as a known biological truth. Thus, dichotomous time-based metrics of the quality of care for children with sepsis, while pragmatic and potentially useful to trend, may be of less value than use of continuous variables such as median time to antimicrobials. Despite a very low quality of evidence on this topic, we provide a strong recommendation because the panel concluded that most patients would accept and most clinicians should seek to initiate antimicrobial therapy as soon as pos-sible after recognition of septic shock in most situations.

For children “without clinical signs of shock,” the panel acknowledged that the diagnosis of sepsis-associated organ dysfunction has additional challenges related to the need to discriminate those with true sepsis from among a large number presenting with suspected infection (44). In view of the avail-able evidence, we suggest starting antimicrobial therapy as soon as possible after sepsis recognition, while allowing up to 3 hours for appropriate diagnostic investigation for patients without clinical signs of shock and for those with an uncer-tain diagnosis. However, the diagnostic evaluation should be performed expeditiously and, if and when the evaluation sup-ports a likely infection or evidence of septic shock or other

sepsis-associated organ dysfunction becomes manifest, anti-microbial therapy should be immediately administered.

7) We recommend empiric broad-spectrum therapy with one or more antimicrobials to cover all likely pathogens (BPS).

8) Once the pathogen(s) and sensitivities are available, we rec-ommend narrowing empiric antimicrobial therapy cov-erage (BPS).

9) If no pathogen is identified, we recommend narrowing or stopping empiric antimicrobial therapy according to clin-ical presentation, site of infection, host risk factors, and ade-quacy of clinical improvement in discussion with infectious disease and/or microbiological expert advice (BPS).

Rationale: Sepsis mortality is associated with delays to “appropriate” antimicrobial therapy, and hence optimal treat-ment for sepsis relies on accurate selection of antimicrobials to ensure activity against the major pathogens (50, 63, 70, 75). “Empiric therapy” refers to the initial choice of antimicrobials pending microbiological results (Table 4) and is based on the predicted likelihood of bacterial pathogens. Empiric therapy should cover a broad range of pathogens that are likely to cause the infection, acknowledging that, in rare circumstances, this may not fully cover very unusual pathogens. “Broad-spectrum therapy” refers to the use of “single- or multi-drug” antimicro-bial therapy with activity against multiple groups of bacteria/pathogens. Broad-spectrum therapy is recommended for in-itial empiric therapy of children with septic shock or sepsis-associated organ dysfunction to increase the likelihood that the initial empirical therapy is effective against the causative pathogens.

The initial choice of empiric antimicrobials should take into account the specific clinical history (e.g., age, site of infection, concomitant disease states, comorbid conditions, indwell-ing devices). Patients with recent or current hospital exposure should receive empiric therapy that considers known infection or colonization, as well as any recent antimicrobial exposure. Institutions or regions should identify the most appropriate first-line single-agent antimicrobial, taking into account ana-tomic site of infection, age, local epidemiology, and host co-morbidity and risk factors (e.g., ceftriaxone is recommended for community-acquired sepsis by the National Institute for Health and Care Excellence [NICE] in the United Kingdom) (16). For complex patients or those recently or currently in hospital, the choice of empiric antimicrobials should also take into account concomitant underlying diseases, chronic organ failure, indwell-ing devices, the presence of immunosuppression or other form of immunocompromise, recent known infection or colonization with specific pathogens, and recent receipt of antimicrobials (65, 76, 77). When available, an infectious diseases clinician should be consulted. Other nonbacterial pathogens that are suspected as a cause of infection should also be targeted as part of initial antimicrobial therapy on a case-by-case basis.

Sepsis in children is most commonly due to gram-nega-tive or gram-positive bacteria, although the relative prevalence of these pathogens varies by age, geographic region, location (community vs hospital) of sepsis onset, and other patient

Page 11: Surviving Sepsis Campaign International Guidelines for the ... · In 2001, the Surviving Sepsis Campaign (SSC) was formed by the Society of Critical Care Medicine (SCCM), European

Copyright © 2020 by the Society of Critical Care Medicine and the World Federation of Pediatric Intensive and Critical Care Societies.Unauthorized reproduction of this article is prohibited

Copyright © 2020 by the Society of Critical Care Medicine and the World Federation of Pediatric Intensive and Critical Care Societies.Unauthorized reproduction of this article is prohibited

Weiss et al

e62 www.pccmjournal.org February 2020 • Volume 21 • Number 2

factors. Invasive fungal infections are largely restricted to im-munocompromised patients and preterm infants. Certain spe-cific conditions put patients at risk for atypical or resistant pathogens, thus requiring specific empiric regimens. For ex-ample, neutropenic patients are at risk for an especially wide range of potential pathogens, including resistant gram-nega-tive bacilli and Candida species, and neonates are at risk for sepsis caused by Listeria monocytogenes and disseminated herpes simplex virus (HSV). Children with chronic conditions treated in hospital settings are prone to sepsis with resistant bacteria such as methicillin-resistant Staphylococcus aureus (MRSA) and vancomycin-resistant enterococci. For children at risk for multidrug-resistant bacterial infections, empiric broad-spectrum antimicrobial regimens may require more than one agent to broadly cover such potential pathogens.

For specific empiric broad-spectrum antimicrobial therapy, the reader is directed to published resources (76, 78) and the need to consider patient history, allergies, local epidemiology, and suspected site/source of infection. However, general sug-gestions can be provided here. For previously healthy children with community-acquired sepsis, a third-generation cephalo-sporin (e.g., ceftriaxone) may be sufficient. Vancomycin should be added in settings where MRSA or ceftriaxone-resistant pneumococci are prevalent, and addition of an aminoglyco-side or substitution of a carbapenem is appropriate in settings where ceftriaxone resistance is common in gram-negative bacteria (79). For immunocompromised patients or hospi-tal-acquired sepsis, antimicrobial therapy should begin with an anti-pseudomonal third- or higher-generation cephalo-sporin (e.g., cefepime), a broad-spectrum carbapenem (e.g., meropenem, imipenem/cilastatin), or an extended-range

penicillin/β-lactamase inhibitor combination (e.g., piperacil-lin/tazobactam) (78). For neonates, therapy should also include ampicillin for listeria and consideration for empiric acyclovir if there is a clinical concern for HSV (76). For patients with a suspected or documented intra-abdominal source of infec-tion, therapy should include broad coverage for gastrointes-tinal pathogens, including anaerobic bacteria, with either an extended-range penicillin/β-lactamase inhibitor combination or carbapenem, or addition of clindamycin or metronidazole. For patients who present with sepsis complicating an influ-enza-like illness during the local influenza season, empiric an-tiviral therapy should be started while awaiting the respiratory virus testing (80, 81). Patients at higher risk of antibiotic-resis-tant infection because of past infection or colonization, local epidemiology, or recent broad-spectrum antibiotic use should receive an individually tailored empiric therapeutic regimen (82). In cases of suspected toxic shock syndrome or necro-tizing fasciitis, empiric treatment should include clindamycin or lincomycin to limit toxin production and enhance bacterial clearance (83). Finally, for sepsis treated in regions endemic for rickettsial or parasitic pathogens (e.g., malaria), clinicians should consider adding relevant empiric coverage.

“Targeted or definitive therapy” refers to the antimicrobial regimen targeted to a specific pathogen(s) after microbio-logic identification. As with empiric therapy, targeted/defini-tive therapy may be single- or multi-drug therapy, but should not be broader than required to treat the specific pathogen(s) after microbiologic identification (84, 85). Risks of unneces-sary continuation of broad-spectrum antibiotic and other antimicrobial therapy include direct side effects and toxicities (such as the nephrotoxicity or ototoxicity of aminoglycosides),

TABLE 4. Definitions for Empiric, Targeted/Definitive, Broad-Spectrum, and Multiple Drug Antimicrobial Therapy

Term Definition Comment

Empiric antimicrobial therapy

Initial antimicrobial therapy started for suspected infection in the absence of definitive microbiologic pathogen identification

Empiric therapy may consist of single or multiple agents but should be broad spectrum in nature. Empiric antimicrobial therapy should be based on local, regional, or national pathogen epidemiology and patient risk factors

Targeted/definitive antimicrobial therapy

Antimicrobial therapy targeted to specific pathogen(s), usually after microbiologic identification

Targeted/definitive therapy may consist of single or multiple agents, but should not be broader than required to treat the specific pathogen(s) after microbiologic identification

Broad-spectrum antimicrobial therapy

An antimicrobial regimen with activity against multiple different groups of bacteria or other pathogens considered to be likely causes of the clinical presentation

Broad-spectrum antimicrobial therapy may consist of single or multiple agents

Multiple-drug antimicrobial therapy

More than one antimicrobial agent is needed to either 1) expand the spectrum of coverage to include additional pathogens (e.g., vancomycin for methicillin-resistant Staphyloccocus aureus; 2) decrease the likelihood of resistance to any particular single agent (e.g., for patients with known or high-risk for multi-drug resistant organism); or 3) provide synergy to treat a suspected or known pathogen

Page 12: Surviving Sepsis Campaign International Guidelines for the ... · In 2001, the Surviving Sepsis Campaign (SSC) was formed by the Society of Critical Care Medicine (SCCM), European

Copyright © 2020 by the Society of Critical Care Medicine and the World Federation of Pediatric Intensive and Critical Care Societies.Unauthorized reproduction of this article is prohibited

Online Special Article

Pediatric Critical Care Medicine www.pccmjournal.org e63

infection with Clostridioides difficile (formerly Clostridium) or fungal pathogens, and promotion of antimicrobial resistance in the patient and in the community. In addition, unnecessary exposure to antibiotics may lead to alteration of the human microbiome early in life, the impact of which is poorly under-stood but has been associated with worse outcomes such as necrotizing enterocolitis in newborns.

Because most microbiological cultures show significant growth within 24 to 36 hours of collection when a pathogen is present (86), empiric treatment should be reevaluated after no more than 48 hours following initiation. If no pathogen is identified and bacterial/fungal infection is deemed unlikely, clinicians should stop empiric antimicrobial therapy to reduce unnecessary exposure to antibiotics/antifungals. However, many children with a clinical diagnosis of septic shock do not have a pathogen isolated (5, 6). Patients with negative bacte-rial microbiological results may have false-negative tests due to antibiotic pretreatment, absence of bacteremia (e.g., bacterial pneumonia despite true bacterial infection), or sepsis-related to viral infections (87). Thus, the decision to continue, narrow, or stop antimicrobial therapy must often be made on the basis of clinician judgment and indirect clinical information, taking into account the clinical presentation, site and type of infec-tion, host risk factors, and adequacy of clinical improvement. Complex patients should be discussed with pediatric infectious diseases and/or microbiology specialists to ensure likely patho-gens are treated and that antibiotics and other antimicrobials are stopped when they are no longer necessary.

10) In children without immune compromise and without high risk for multidrug-resistant pathogens, we suggest against the routine use of empiric multiple antimicrobials directed against the same pathogen for the purpose of syn-ergy (weak recommendation, very low quality of evidence).

Remarks: In certain situations, such as confirmed or strongly suspected group B streptococcal sepsis, use of empiric multiple antimicrobials directed against the same pathogen for the purpose of synergy may be indicated.

11) In children with immune compromise and/or at high risk for multidrug-resistant pathogens, we suggest using empiric multi-drug therapy when septic shock or other sepsis-associated organ dysfunction is present/suspected (weak recommendation, very low quality of evidence).

Rationale: The selection of an empiric antimicrobial reg-imen requires consideration of a patient’s underlying disease state, potential history of prior infections and colonization with multidrug-resistant organisms, presence of immunosuppres-sion, and possible recent antimicrobial use, as well as local path-ogen prevalence and susceptibility profile (50, 88, 89). Empiric therapy may be single- or multi-drug, but should be broad spec-trum in nature as defined in Table 4. For select patients or with concern for particular types of infection, this may necessitate adding a glycopeptide (i.e., vancomycin) to ensure empiric cov-erage of MRSA or a second gram-negative agent (e.g., aminogly-coside in addition to a beta-lactam or second/third-generation

cephalosporin) when antibiotic resistance is a concern. How-ever, routinely including an aminoglycoside or a glycopeptide for synergy or “double-coverage” as part of an empiric regimen is not supported by the available data (89–100).

A recent Cochrane review evaluated beta-lactam mono-therapy versus beta-lactam and aminoglycoside combination regimens for sepsis and included 69 trials accounting for 7,863 participants, including neonatal and pediatric patients (88). In trials where the mono- and multidrug arm used the same beta-lactam, no difference in clinical outcomes was observed between study groups. In studies where the monotherapy arm contained a beta-lactam of broader spectrum than the multi-drug arm, monotherapy showed a possible benefit for all-cause mortality (OR, 0.85; 95% CI, 0.71–1.01) and a significant ad-vantage for clinical failure (OR, 0.75; 95% CI, 0.67–0.84) (88). Additionally, indirect evidence from adults with sepsis including 13 RCTs comparing empirical mono- versus com-bination antibiotic therapy suggests mortality and other out-comes are not improved by empiric combination therapy (90). Therefore, many children with septic shock and other sepsis-associated organ dysfunction do not require empiric multi-drug therapy. Clinicians should continually reevaluate the local epidemiology and resistance rates to ensure monotherapy re-mains appropriate (88).

Certain clinical scenarios, however, may necessitate multi-drug antimicrobial therapy. For example, in patients at high risk for resistant gram-negative infections with sepsis, com-bining a beta-lactam/beta-lactamase inhibitor agent (i.e., piperacillin/tazobactam combination) with an aminogly-coside (i.e., gentamicin) can be considered, not for synergy, but for expanded coverage to treat both susceptible and re-sistant pathogens until final identification and susceptibilities are known (101–103). Additionally, a synergistic multi-drug regimen may be appropriate in select settings, even for tar-geted/definitive therapy, such as device-associated infec-tions, enterococcal endocarditis, staphylococcal endocarditis, group B streptococcal sepsis, and carbapenem-resistant Enterobacteriaceae infections (104, 105).

Pediatric patients with cancer and transplant recipients have a substantial degree of immunosuppression and repre-sent a population at higher risk for colonization and infec-tion with multi-drug resistant organisms (106, 107). The 2017 guidelines for the management of fever and neutropenia (FN) in children with cancer and hematopoietic stem-cell trans-plantation recommended monotherapy with an anti-pseudo-monas beta-lactam, a fourth-generation cephalosporin, or a carbapenem as empiric therapy in high-risk pediatric patients with FN (78). The three RCTs in high-risk pediatric FN com-paring monotherapy with aminoglycoside-containing com-bination therapy found no significant differences in failure rates, infection-related mortality, or overall mortality (78, 108, 109). The meta-analysis also confirmed the efficacy and safety of monotherapy without the addition of an aminoglycoside. However, the 2017 guidelines on the management of children with FN did recommend addition of a second gram-negative agent and/or a glycopeptide when resistant organisms were

Page 13: Surviving Sepsis Campaign International Guidelines for the ... · In 2001, the Surviving Sepsis Campaign (SSC) was formed by the Society of Critical Care Medicine (SCCM), European

Copyright © 2020 by the Society of Critical Care Medicine and the World Federation of Pediatric Intensive and Critical Care Societies.Unauthorized reproduction of this article is prohibited

Copyright © 2020 by the Society of Critical Care Medicine and the World Federation of Pediatric Intensive and Critical Care Societies.Unauthorized reproduction of this article is prohibited

Weiss et al

e64 www.pccmjournal.org February 2020 • Volume 21 • Number 2

suspected for patients who are clinically unstable (i.e., septic shock) and in centers with a high rate of resistant pathogens (78). Therefore, for children with septic shock or other sepsis-associated organ dysfunction who have immune compromise and/or are at high risk for multidrug-resistant pathogens, we suggest empiric multi-drug therapy.

Currently, specific resistance rate thresholds do not exist to help clinicians decide when the addition of a glycopeptide or second gram-negative agent for sepsis or septic shock is neces-sary. The U.S. guidelines for the management of community-acquired pneumonia in adults suggest a 25% rate of high-level macrolide resistance in the community as the threshold beyond which macrolides should not be used (110, 111). Additionally, current guidelines from the Infectious Diseases Society of America recommend an alternative antibiotic for skin and soft tissue infections if the local clindamycin resistance rate is greater than 10% (112). Considering the current rates of mor-bidity and mortality for patients with sepsis or septic shock, a local or regional antimicrobial resistance rate exceeding 10% is probably a prudent threshold for the addition of a second agent if that pathogen is suspected (5, 63).

12) We recommend using antimicrobial dosing strategies that have been optimized based on published pharmacokinetic/pharmacodynamic principles and with consideration of specific drug properties (BPS).

Rationale: Sepsis may alter the pharmacokinetics and phar-macodynamics of antimicrobials. Therefore, antimicrobial dosing should be individualized to deliver effective and timely treatment of life-threatening infection, while at the same time limiting adverse medication effects. Sub-therapeutic dosing can lead to failure to clear the infection, prolong organ dys-function, and can lead to the development of antimicrobial resistance. A substantial proportion of sepsis patients are at risk for altered drug metabolism and/or clearance, including those with kidney and hepatic dysfunction and those treated with extracorporeal therapies (113). In particular, continuous renal replacement therapy (CRRT) and extracorporeal mem-brane oxygenation (ECMO) both lead to profound alteration of antimicrobial clearance, requiring individual dose adapta-tion (114). Therapeutic drug monitoring, where available, can permit individualized antimicrobial dosing to achieve max-imal effect while minimizing toxicity (115).

Examples of sepsis and septic shock-related altered pharma-cokinetics include increased volume of distribution as a result of fluid therapy and capillary leak (116), decreased antimi-crobial clearance as a result of altered renal and hepatic organ perfusion and organ dysfunction (117), and higher unbound drug levels due to hypoalbuminemia leading to increased clearance(118). Hepatic dysfunction impairs the metabolism of lipophilic and highly albumin-bound antibiotics, leading to drug accumulation and toxicity. In renal dysfunction, time-dependent antibiotics cleared by the kidneys, such as the beta-lactams, require reduced dosing frequency.

The three main determinants of antimicrobial efficacy are 1) the time during which the concentration of the drug

remains above the minimum inhibitory concentration (MIC) of the causative pathogen (T > MIC) (time-dependent anti-biotics); 2) the peak concentration to MIC ratio (C

max/MIC)

(concentration-dependent antibiotics); and 3) the ratio of the 24-hour area under the concentration-time curve di-vided by the MIC (AUC24/MIC) (concentration-dependent with time-dependence antibiotics). The main classes of time-dependent antibiotics include beta-lactams (penicillins, cephalosporins, carbapenems, monobactams) and lincos-amides (clindamycin and lincomycin). For amoxicillin-cla-vulanic acid, current published dosing regimens in critically ill children can result in sub-therapeutic concentrations in the early period of sepsis due to augmented renal clearance (119, 120). In sepsis, the use of continuous or extended infu-sions with loading doses, as opposed to intermittent dosing, may lead to improved outcomes in patients treated with beta-lactam antibiotics (121).

The main classes of concentration-dependent antibiotics include aminoglycosides and metronidazole. In some centers, drug concentrations measured within 60 minutes before or after administration of aminoglycosides are used to estimate the C

min and C

max, respectively, and together with the MIC of

the pathogen, can help to guide appropriate antimicrobial dos-ing (118). Concentration-dependent antibiotics may require an altered dosing frequency to maximize bacterial killing by preserving the C

max/MIC.

Glycopeptides, oxazolidinones, fluoroquinolones, polymix-ins, daptomycin, azithromycin, and tigecycline are examples of concentration-dependent with time-dependent antibiotics. For vancomycin, this can mean higher doses, but that comes with an increased risk of toxicity. For this reason, continuous vancomycin infusions may be considered to achieve optimal concentrations in some patients (122). For concentration-dependent with time-dependent antibiotics, dose optimization involves adjusting the dosing interval rather than administered dose (118).

13) In children with septic shock or sepsis-associated organ dysfunction who are receiving antimicrobials, we recom-mend daily assessment (e.g., clinical, laboratory assess-ment) for de-escalation of antimicrobial therapy (BPS).

Remarks: This assessment should include a review of the ongoing indication for empiric antimicrobial therapy after the first 48 hours that is guided by microbiologic results and in response to clinical improvement and/or evidence of infection resolution. This recommendation applies to patients being treated with empiric, targeted, and combination therapy.

Rationale: The misuse and overuse of broad-spectrum antimicrobials in healthcare, the community, veterinary medi-cine, and the environment have contributed to a global public health emergency (123). De-escalation of antimicrobials, where appropriate, is warranted to minimize adverse effects of unnecessarily prolonged administration. To date, QI efforts in adults have shown that safe and effective antimicrobial de-es-calation can be achieved by daily assessment and discussion (124, 125).

Page 14: Surviving Sepsis Campaign International Guidelines for the ... · In 2001, the Surviving Sepsis Campaign (SSC) was formed by the Society of Critical Care Medicine (SCCM), European

Copyright © 2020 by the Society of Critical Care Medicine and the World Federation of Pediatric Intensive and Critical Care Societies.Unauthorized reproduction of this article is prohibited

Online Special Article

Pediatric Critical Care Medicine www.pccmjournal.org e65

Several host biomarkers have also been proposed to aid in the safe de-escalation of antimicrobial therapy. In adults with severe infections and sepsis, procalcitonin has been shown to successfully guide de-escalation (126–130) with an associ-ated improved mortality (131). Similar reductions in length of antimicrobial therapy have also been safely achieved in ne-onatal populations (132) using procalcitonin as a guide. In the United Kingdom, the NICE committee concluded that in emergency department and critical care settings, procalcito-nin testing shows promise, but currently, insufficient evidence is available to recommend the routine adoption of procalci-tonin-guided antimicrobial de-escalation (www.nice.org.uk/guidance/dg18).

Although a relationship between antimicrobial steward-ship programs (ASPs) and a decrease in antimicrobial resist-ance has not yet been shown, studies suggest that inpatient pediatric ASPs may reduce antimicrobial usage without con-tributing to adverse patient outcomes (123). The “Start Smart - Then Focus” work from Public Health England suggests a pragmatic approach of the five “antimicrobial prescribing de-cision” options to include 1) stop antimicrobials if there is no evidence of infection, 2) switch antimicrobials from IV to oral, 3) change antimicrobials—ideally to a narrower spectrum—or broader if required, 4) continue and document next review date or stop date, and 5) outpatient parenteral antimicrobial therapy (133). De-escalating antimicrobial therapy must be based in sound clinical judgment and needs to be adapted to local epidemiology and identified resistance patterns.

14) We recommend determining the duration of antimicrobial therapy according to the site of infection, microbial eti-ology, response to treatment, and ability to achieve source control (BPS).

Rationale: The main purposes of antimicrobial therapy in patients with sepsis are to reduce the pathogen load rapidly and to prevent recurrence. Important determinants of the re-quired duration of antimicrobial therapy include site of infec-tion, ability to drain or remove fixed infectious foci, choice of antimicrobial therapy, time to clearance of positive cultures, the nature of the causative pathogen, and the integrity of the host immune response. There is no evidence that severity of sepsis is an important determinant of optimal duration of therapy because illness severity is not expected to affect clear-ance of infection.

The optimal duration of antimicrobial therapy can differ by site of infection because of a high pathogen burden, poor antimicrobial penetration, or presence of difficult-to-eradicate microbial biofilms at the site. For example, longer duration of therapy is typically required for treatment of endocarditis, un-drained abscesses, and prosthetic joint infection without device removal (134–136). Characteristics of the causative organism that may affect optimal duration of therapy include resistance or decreased susceptibility to front-line antimicrobials and propensity to cause deep-seated or difficult-to-eradicate infec-tion. For example, optimal duration of treatment for endocar-ditis caused by methicillin-susceptible S. aureus may be shorter

than for that caused by MRSA (136). Similarly, although 7–10 days of therapy is appropriate for treatment of uncomplicated gram-negative bacteremia in immunocompetent hosts (137, 138), uncomplicated S. aureus bacteremia requires a longer course of therapy to effect cure (139–141), likely because of unrecognized seeding (142). Integrity of host immunity may also affect clearance of infection, so antimicrobial therapy for infection in neutropenic pediatric patients with cancer is often continued until resolution of neutropenia (78).

A systematic review evaluated studies describing duration of treatment for clinically and microbiologically-documented infections in children and provides evidence-based clinical guidelines for optimal duration of antimicrobial therapy for specific conditions (143). Given the lack of studies on the du-ration of antimicrobial therapy for pediatric patients with sepsis specifically, we refer to this previously published guide-line as best evidence. Importantly, there are no data to support that the presence of organ dysfunction or a higher initial illness severity necessitates longer therapy for specific infection types (other than attention to how such organ dysfunction may affect antimicrobial pharmacokinetics and pharmacodynamics).

Observational studies suggest that longer exposure to antibiotics is associated with risk of potential adverse events including necrotizing enterocolitis in very low birthweight infants (144), candidemia in hospitalized children (145, 146), development of antimicrobial resistance (147) and C. difficile (formerly Clostridium) infection (148). Several meta-analyses, RCTs, and observational studies have compared long- versus short-duration antibiotic therapy for serious infections (140, 144, 149–167). Most studies suggest that shorter courses were associated with similar clinical outcomes compared with longer durations; these include neonatal bacteremia (158, 163), pyelo-nephritis (168), uncomplicated bacterial meningitis (154, 155, 159–161, 164, 165), and pneumonia (169, 170). In contrast to these infections, some studies have identified scenarios where longer durations of antimicrobial therapy is superior. For ex-ample, an RCT suggested that 14 days of antibiotic therapy was superior to 7 days for treatment of neonates with S. aureus bacteremia (140), and an observational study suggested that greater than 10 days was superior to less than or equal to 10 days of antibiotic therapy in children treated for gram-nega-tive bacteremia without removal of a preexisting central ve-nous catheter (CVC) (162).

SOURCE CONTROL

15) We recommend that emergent source control intervention be implemented as soon possible after a diagnosis of an infection amenable to a source control procedure is made (BPS).

Remarks: Appropriate diagnostic testing to identify the site of infection and microbial etiology should be performed, and advice from specialist teams (e.g., infectious diseases, surgery) should be sought, as appropriate, in order to prioritize inter-ventions needed to achieve source control.

Page 15: Surviving Sepsis Campaign International Guidelines for the ... · In 2001, the Surviving Sepsis Campaign (SSC) was formed by the Society of Critical Care Medicine (SCCM), European

Copyright © 2020 by the Society of Critical Care Medicine and the World Federation of Pediatric Intensive and Critical Care Societies.Unauthorized reproduction of this article is prohibited

Copyright © 2020 by the Society of Critical Care Medicine and the World Federation of Pediatric Intensive and Critical Care Societies.Unauthorized reproduction of this article is prohibited

Weiss et al

e66 www.pccmjournal.org February 2020 • Volume 21 • Number 2

16) We recommend removal of intravascular access devices that are confirmed to be the source of sepsis or septic shock after other vascular access has been established and depending on the pathogen and the risks/benefits of a sur-gical procedure (strong recommendation, low quality of evidence).

Rationale: Source control is defined as physical modalities taken to control or remove the source of infection or to prevent spread of the infection systemically or to adjacent tissues (171). Source control may include percutaneous or deep abscess drainage, drainage of an empyema, septic joint, or subperi-osteal abscess, removal of infected hardware or CVCs, or de-bridement of necrotizing soft-tissue infection. The adult SSC guidelines recommend source control as soon as is reasonably feasible after resuscitation, ideally within 6–12 hours of diag-nosis (50). Waiting for patients to clinically stabilize prior to intervention is not recommended, as delaying adequate source control may lead to further clinical deterioration (6). Although source control as an adjunct to antimicrobial and other med-ical therapy has been best described for abdominal infections in adults and has been associated with reduction in mortality (172), the role of source control for pediatric sepsis has been less well elucidated (173).

The importance of source control in children has been shown for skin and deep tissue abscesses and necrotizing fas-ciitis (173–175). Despite the relative paucity of pediatric data, source control is an important facet of treatment of sepsis, and should not be delayed. Larger collections containing infected material often are poorly penetrated by IV antimicrobials and contribute to direct and hematogenous spread, ongoing in-flammation, and organ dysfunction.

A common, but potentially preventable, source of infection is central line-associated bloodstream infections. Delayed re-moval of a CVC in neonates and in patients with fungemia or Enterobacteriaceae bacteremia increases the risk of death or slows recovery (176–179). Removal of a CVC that is the source of infection is therefore generally warranted unless extenuat-ing circumstances exist. Fungal infection dictates immediate removal, while in case of coagulase-negative Staphylococcus species or clinically stable patients with infection caused by gram-negative bacilli, infections can often be initially treated through the CVC as a temporizing measure. The decision to remove the CVC, or not, should ultimately be made based on the pathogen suspected/recovered and host factors, such as immune status (Supplemental Table 3, Supplemental Digital Content 1, http://links.lww.com/PCC/B138).

FLUID THERAPY

17) In healthcare systems with availability of intensive care, we suggest administering up to 40–60 mL/kg in bolus fluid (10–20 mL/kg per bolus) over the first hour, titrated to clin-ical markers of cardiac output and discontinued if signs of fluid overload develop, for the initial resuscitation of chil-dren with septic shock or other sepsis-associated organ dys-function (weak recommendation, low quality of evidence).

18) In healthcare systems with no availability of intensive care and in the absence of hypotension, we recommend against bolus fluid administration while starting main-tenance fluids (strong recommendation, high quality of evidence).

19) In healthcare systems with no availability of intensive care, if hypotension is present, we suggest administering up to 40 mL/kg in bolus fluid (10–20 mL/kg per bolus) over the first hour with titration to clinical markers of cardiac output and discontinued if signs of fluid overload develop (weak recommendation, low quality of evidence).

Remarks: Clinical markers of cardiac output may include heart rate, blood pressure, capillary refill time, level of con-sciousness, and urine output. In all settings, the need for fluid administration should be guided by frequent reassessment of clinical markers of cardiac output, serial blood lactate meas-urement and advanced monitoring, when available. Signs of fluid overload that should limit further fluid bolus therapy may include clinical signs of pulmonary edema or new or worsening hepatomegaly.

Rationale: Effective fluid resuscitation in septic shock can correct hypovolemia caused by capillary leak, vasodilation, and fluid losses. Without maintenance of adequate atrial filling pressures, cardiac output will fall and organ perfusion will be compromised.

Three RCTs of different volume resuscitation strategies in children with septic shock in settings in which advanced sup-portive care (e.g., intubation, mechanical ventilation, and in-tensive care) was accessible have been published (180–182). These studies have a combined total of only 316 children and showed no difference in mortality between the restrictive and liberal fluid resuscitation groups (Supplemental Table 4 and Supplemental Fig. 2, Supplemental Digital Content 1, http://links.lww.com/PCC/B138).

In geographic settings in which advanced supportive care, including mechanical ventilation, is limited and/or in-tensive care is not routinely accessible, the only large-scale RCT of different bolus fluid volume resuscitation strate-gies in severe infection in children was the Fluid Expansion as Supportive Therapy (FEAST) study (Supplemental Table 4 and Supplemental Fig. 2, Supplemental Digital Content 1, http://links.lww.com/PCC/B138) (183). The FEAST study was conducted in Africa in a low-resource setting without access to PICU admission. Children between 60 days and 12 years old with a severe febrile illness and abnormal perfusion were ran-domized to either rapid volume expansion with 20 mL/kg of IV 0.9% saline or 5% albumin or no bolus with maintenance fluid only (control group). Among the 3,141 study partici-pants, malaria and anemia were highly prevalent. Overall, the RCT demonstrated a lower mortality after 48 hours in children receiving conservative fluid therapy (i.e., no bolus fluid, main-tenance fluid only) than among those given liberal initial fluid therapy (i.e., 20 mL/kg fluid bolus with maintenance fluid) with a RR of 0.72 (95% CI, 0.57–0.9). Notably, 29 additional children enrolled with severe hypotension (systolic blood pres-sure of < 50 mm Hg in children younger than 12 months old,

Page 16: Surviving Sepsis Campaign International Guidelines for the ... · In 2001, the Surviving Sepsis Campaign (SSC) was formed by the Society of Critical Care Medicine (SCCM), European

Copyright © 2020 by the Society of Critical Care Medicine and the World Federation of Pediatric Intensive and Critical Care Societies.Unauthorized reproduction of this article is prohibited

Online Special Article

Pediatric Critical Care Medicine www.pccmjournal.org e67

< 60 mm Hg in children 1 to 5 years old, and < 70 mm Hg in children older than 5 years old) were treated with 40 mL/kg fluid bolus per the planned protocol without randomization to the control group. One additional child who was randomized to the control group also received a 40 mL/kg fluid bolus due to severe hypotension.

For children with septic shock diagnosed by abnormal per-fusion or hypotension in healthcare systems with availability of advanced supportive and intensive care, and in the absence of signs of fluid overload, the panel suggests administering up to 40–60 mL/kg fluid bolus therapy in the first hour of resuscita-tion. Fluid resuscitation should be titrated to clinical markers of cardiac output and discontinued if signs of fluid overload develop. Clinical markers of cardiac output can include heart rate, capillary refill, and urine output. Although no high-qual-ity RCTs demonstrate clear superiority of this practice, nu-merous observational studies have reported improved patient outcomes with routine administration of up to 40–60 mL/kg fluid bolus therapy in the first hour of resuscitation (8, 32, 33, 36, 184–187). The panel provides only a weak recommenda-tion for this resuscitation strategy in healthcare systems with availability of intensive care because a more restrictive fluid re-suscitation strategy has not been shown to be inferior in this setting and indirect data (183) indicate harm from rapid fluid boluses in other settings. For this recommendation, the panel judged the balance of observational data supporting initial fluid bolus therapy to outweigh an indirect suggestion of harm because the generalizability of the FEAST trial to healthcare systems with availability of advanced supportive and intensive care is not clear.

For children with septic shock without signs of fluid over-load in low-resource settings where advanced supportive and intensive care is not available, the panel recommends against bolus fluid administration, while starting maintenance flu-ids, in the first hour if hypotension is not present, and suggests administering up to 40 mL/kg in bolus fluid (10–20 mL/kg per bolus) over the first hour if hypotension is present. The strong recommendation against bolus fluid if hypotension is not pre-sent was based on the FEAST trial, in which rapid bolus fluid in the first hour of resuscitation increased mortality compared with maintenance fluids only.

For the subset of children with septic shock and hypoten-sion, we suggest cautious administration of fluid bolus therapy in low-resource settings because there are insufficient data to conclude that fluid resuscitation is not beneficial in children with septic shock and hypotension. In the FEAST study, all children with “severe” hypotension were treated with 40 mL/kg of bolus fluid (183), and so it is not known if fluid bolus therapy was beneficial or harmful in this subgroup of chil-dren. It should also be noted that children with gastroenter-itis were excluded from FEAST, as ongoing fluid losses should be replaced with IV or oral rehydration as indicated. A recent analysis of children with “moderate” hypotension who were randomized to either fluid bolus or maintenance fluid in the FEAST trial was published after completion of our initial sys-tematic review but considered by the panel to be potentially

influential (188). In this analysis, only children with moderate hypotension were included because children with severe hy-potension were not allocated to the control (no bolus) arm. Fluid bolus therapy in children with moderate hypotension was not beneficial or harmful compared with maintenance fluid only (RR of death = 1.48; 95% CI, 0.61–3.66; p = 0.41). Although children who were reclassified as meeting all three World Health Organization (WHO) shock criteria of cold extremities, prolonged capillary refill greater than 3 seconds, and weak, fast pulse (14) had 48% mortality in the bolus groups versus 20% mortality in the control group, this dif-ference was not statistically significant (p = 0.07). These cases were a very small proportion of the total FEAST trial partici-pants (only 72 [2.3%] had moderate hypotension and 65 [2%] met the full WHO shock criteria), and no data were provided about differential patient characteristics between these very small post hoc subgroups to assess for potential confounding. Therefore, until further data are available, the panel suggests cautious administration of fluid bolus therapy for the subset of children with septic shock and hypotension in low-resource settings as a weak recommendation based on low quality of evidence.

Although a suggestion of “up to” 40 mL/kg was included for hypotensive shock in low-resource settings because this volume was administered to children with severe hypoten-sion in the FEAST study, fluid administration should always be titrated to clinical markers of cardiac output and discon-tinued if signs of fluid overload develop. For purposes of this weak recommendation, hypotension can be defined as 1) sys-tolic blood pressure of less than 50 mm Hg in children younger than 12 months old, less than 60 mm Hg in children 1 to 5 years old, and less than 70 mm Hg in children older than 5 years old (183) or 2) by the WHO criteria of cold extremities “with” prolonged capillary refill greater than 3 seconds and weak, fast pulse (14). Although the panel did not review differ-ent approaches to fluid bolus therapy in hypotensive children in low-resource settings, WHO recommends 10–20 mL/kg of isotonic crystalloid over 30–60 minutes, followed by an addi-tional 10 mL/kg over 30 minutes if condition has not improved and signs of fluid overload, cardiac failure, or neurologic dete-rioration have not developed (14).

Fluid boluses may be administered as 10 or 20 mL/kg, according to clinician preference. To facilitate rapid IV fluid administration (as well as other IV therapies, such as antimi-crobials and vasoactive medications), clinicians should con-sider alternative methods of vascular access if initial attempts at peripheral vein cannulation are not immediately successful. Intraosseous access is rapid and effective and recommended by Pediatric Advanced Life Support, Advanced Pediatric Life Support, and the International Liaison Committee on Resuscitation. Ultrasound-guided peripheral IV catheter placement, CVCs, and umbilical venous catheter access are alternatives if the skills are immediately available (189, 190). In all healthcare systems, repeat boluses should only be admin-istered after reassessment of hemodynamic status if shock has not resolved and signs of fluid overload are not present.

Page 17: Surviving Sepsis Campaign International Guidelines for the ... · In 2001, the Surviving Sepsis Campaign (SSC) was formed by the Society of Critical Care Medicine (SCCM), European

Copyright © 2020 by the Society of Critical Care Medicine and the World Federation of Pediatric Intensive and Critical Care Societies.Unauthorized reproduction of this article is prohibited

Copyright © 2020 by the Society of Critical Care Medicine and the World Federation of Pediatric Intensive and Critical Care Societies.Unauthorized reproduction of this article is prohibited

Weiss et al

e68 www.pccmjournal.org February 2020 • Volume 21 • Number 2

Although fluid bolus therapy should be discontinued if signs of fluid overload are present or develop, early recognition of fluid overload by clinical examination is a challenge in chil-dren. Identifying fluid overload is especially difficult in young children, in whom crackles (rales) are often absent even in the context of gross pulmonary edema. Worsening respiratory status, particularly increasing respiratory rate, radiographic evidence of pulmonary edema in an intubated patient, or new or expanding hepatomegaly may be the only clues of evolv-ing fluid overload. Bedside ultrasound may also be helpful to assess fluid overload, as there is emerging evidence to suggest that a “full” inferior vena cava with minimal variation across the respiratory cycle demonstrated on ultrasound indicates a fluid-replete circulation (191).

20) We suggest using crystalloids, rather than albumin, for the initial resuscitation of children with septic shock or other sepsis-associated organ dysfunction (weak recommenda-tion, moderate quality of evidence).

Remarks: Although there is no difference in outcomes, this recommendation takes into consideration cost and other bar-riers of administering albumin compared with crystalloids.

Rationale: The FEAST trial investigated 3,141 African chil-dren with infection and impaired perfusion, who were ran-domly assigned to resuscitation with 5% human albumin solution or 0.9% saline boluses or no boluses at admission to the hospital. Although both the albumin and 0.9% saline arms exhibited higher mortality than the no bolus arm, comparing human albumin solution to 0.9% saline (RR, 1.02; 95% CI, 0.8–1.28) showed no difference in mortality (183). In the absence of any clear benefit of albumin administration in children with sepsis, and in view of the additional costs in comparison to crystalloids, problems of availability, and the potential risk of blood-borne infection, we suggest against the routine use of albumin for initial fluid resuscitation in children with sepsis.

21) We suggest using balanced/buffered crystalloids, rather than 0.9% saline, for the initial resuscitation of children with septic shock or other sepsis-associated organ dysfunc-tion (weak recommendation, very low quality of evidence).

Rationale: Increasing evidence from observational stud-ies and RCTs in adults suggests that resuscitation with crys-talloid fluids containing high chloride concentrations (e.g., 0.9% saline) is associated with hyperchloremic acidosis, sys-temic inflammation, acute kidney injury (AKI), coagulopathy, and mortality when compared with resuscitation with more balanced/buffered crystalloids (e.g., lactated Ringer’s, Plasma-Lyte) (192). Although no pediatric RCTs compare balanced/buffered crystalloids to 0.9% saline, there are two large obser-vational studies in children with sepsis (193, 194). They in-cluded a total of 30,532 children with sepsis, 2,100 of whom received only balanced/buffered crystalloids for the first 72 hours of hospital admission, and 28,432 who received 0.9% saline (Supplemental Table 5, Supplemental Digital Content 1, http://links.lww.com/PCC/B138). These studies showed that use of balanced/buffered crystalloids was associated with lower

mortality (OR, 0.79; 95% CI, 0.65–0.95) but not AKI (OR, 0.98; 95% CI, 0.94–1.02) (193, 194). Indirect evidence from adult patients, including two large RCTs, also demonstrates ben-efit with balanced/buffered crystalloids over 0.9% saline, with adult patients who received larger volumes of fluid and those with sepsis exhibiting the greatest benefit (192, 195). Taken to-gether, these data support that the desirable consequences of balanced/buffered crystalloids probably outweigh the unde-sirable consequences (including cost), especially in those who require large volume of fluid resuscitation. Therefore, pending further high-quality pediatric data, we suggest that balanced/buffered crystalloids should generally be preferred over 0.9% saline for resuscitation of children with septic shock or other sepsis-associated organ dysfunction without a specific indica-tion for an alternative fluid type (e.g., 0.9% saline may be pre-ferred in patients with hyponatremia or concern for increased intracranial pressure).

22) We recommend against using starches in the acute re-suscitation of children with septic shock or other sepsis-associated organ dysfunction (strong recommendation, moderate quality of evidence).

Rationale: No studies compare starches with other fluids in children. However, in adults with severe sepsis and septic shock (Supplemental Table 6, Supplemental Digital Content 1, http://links.lww.com/PCC/B138), two large RCTs showed increased risk of mortality, coagulopathy, and AKI in patients receiving hydroxyethyl starch (HES) (196, 197). A meta-analy-sis further confirmed the risk of harm with HES (198). In the United States, the Food and Drug Administration has restricted the use of HES (199) and the European Medicines Agency has recommended complete suspension of its use (200). There-fore, we strongly recommend against the use of HES in chil-dren with sepsis.

23) We suggest against using gelatin in the resuscitation of children with septic shock or other sepsis-associated organ dysfunction (weak recommendation, low quality of evidence).

Rationale: One RCT of gelatin-derived fluid in pediatric septic shock compared it to 0.9% saline in 60 patients. The esti-mates were imprecise, and showed no difference in mortality, days of using vasoactive medications, or AKI between the two groups (201) (Supplemental Table 7, Supplemental Digital Content 1, http://links.lww.com/PCC/B138). In the absence of any data indicating benefit of gelatin in children, we suggest against its use in pediatric sepsis.

HEMODYNAMIC MONITORING

24) We were unable to issue a recommendation about whether to target mean arterial blood pressure (MAP) at the 5th or 50th percentile for age in children with septic shock and other sepsis-associated organ dysfunction. However, in our practice, we target MAP to between the 5th and 50th per-centile or greater than 50th percentile for age.

Page 18: Surviving Sepsis Campaign International Guidelines for the ... · In 2001, the Surviving Sepsis Campaign (SSC) was formed by the Society of Critical Care Medicine (SCCM), European

Copyright © 2020 by the Society of Critical Care Medicine and the World Federation of Pediatric Intensive and Critical Care Societies.Unauthorized reproduction of this article is prohibited

Online Special Article

Pediatric Critical Care Medicine www.pccmjournal.org e69

Rationale: Although no data from RCTs support specific hemodynamic targets in children, evidence suggests that tar-geting MAP of approximately 65 mm Hg (5th percentile) in adults with septic shock may be beneficial (202). In the absence of evidence from RCTs, we were unable to reach consensus to recommend a specific MAP target for children. However, in our practice, 37% of panel members reported targeting MAP between the 5th and 50th percentile for age and 45% reported targeting MAP greater than 50th percentile for age. Many panelists also commented that lower blood pressures are ac-ceptable if other hemodynamic variables (e.g., mental status, perfusion, urine output, lactate) are improving. RCTs to define optimal hemodynamic targets, including MAP, are urgently re-quired to inform practice in pediatric sepsis. In settings where direct measurement of MAP is less reliable, systolic blood pres-sure provides a reasonable alternative.

A previous recommendation to target perfusion pressure (MAP minus central venous pressure [CVP]) lacks supporting data (203). Prioritizing CVP measurement is also impractical during early resuscitation (such as in most pediatric emer-gency departments); CVP also provides an unreliable assess-ment of left ventricular preload.

25) We suggest not using bedside clinical signs in isolation to categorize septic shock in children as “warm” or “cold” (weak recommendation, very low quality of evidence).

26) We suggest using advanced hemodynamic variables, when available, in addition to bedside clinical variables to guide the resuscitation of children with septic shock or other sep-sis-associated organ dysfunction (weak recommendation, low quality of evidence).

Remarks: Advanced hemodynamic monitoring may in-clude cardiac output/cardiac index, systemic vascular resist-ance, or central venous oxygen saturation (Scvo

2).

Rationale: The ACCM previously recommended clinical assessment of children in septic shock to differentiate “warm” versus “cold” shock based on extremity temperature, capillary refill, pulse strength, diastolic blood pressure, and pulse pres-sure. Depending on “warm” or “cold” classification, different resuscitation strategies were suggested (e.g., fluid and vaso-pressors for “warm” shock and inotropes for “cold” shock). However, a number of observational studies have demon-strated very poor correlation of clinical assessments with car-diac index and systemic vascular resistance as measured by advanced monitoring (204–209). Indeed, many children who appeared to have “warm” shock by clinical examination had evidence of myocardial dysfunction, thus demonstrating the challenge of using clinical signs alone to direct therapy. Hence, we suggest not attempting to make this distinction using clin-ical assessments alone, although this categorical distinction may be helpful if advanced hemodynamic monitoring is avail-able to assess patient physiology more accurately. Examples of advanced monitoring include invasive arterial blood pressure monitoring with pulse contour analysis, ultrasound Doppler of the ascending or descending thoracic aorta (suprasternal or esophageal Doppler), cardiac ultrasound/echocardiography

(210), or measurement of Scvo2 (190). All of these variables

(other than Scvo2) will provide additional assessment of car-

diac index and/or systemic vascular resistance index beyond clinical signs, which may then be used to direct and titrate treatment. There is also emerging evidence that fluid respon-siveness may be predicted by aortic blood flow peak velocity variation in mechanically ventilated children (211). In an RCT of 90 children admitted to a PICU in Egypt, addition of serial echocardiography provided early recognition of septic myo-cardial dysfunction and hypovolemia that was not apparent on clinical assessment and resulted in faster shock reversal, less fluid overload, shorter LOS, and lower mortality compared with the group without serial echocardiography (210). When advanced hemodynamic monitoring is available, it is appro-priate to target the normal range for variables such as cardiac index, systemic vascular resistance index, stroke index, and Scvo

2 (Table 5). No evidence supports targeting a supranormal

range of cardiac index.Until recently, adult guidelines have recommended early

goal-directed therapy (EGDT) based on the protocol published by Rivers et al (212). This recommendation described the use of a series of “goals” that included CVP and Scvo

2. This approach

is no longer recommended following a failure to show reduc-tion in mortality in three subsequent large multicenter RCTs (213–215). In children, there has only been one small RCT sup-porting the use of a protocolized approach including targeting Scvo

2 greater than 70%. This study included 102 children with

fluid-refractory septic shock (RSS) and showed a reduced risk of death (RR, 0.3; 95% CI, 0.13–0.68) from a very high base-line mortality of 39% (26). No high-quality RCTs have investi-gated other hemodynamic variables to guide therapy in children (Supplemental Table 8 and Supplemental Fig. 3, Supplemental Digital Content 1, http://links.lww.com/PCC/B138).

27) We suggest using trends in blood lactate levels, in addi-tion to clinical assessment, to guide resuscitation of chil-dren with septic shock and other sepsis-associated organ dysfunction (weak recommendation, very low quality of evidence).

Remarks: In children with an elevated blood lactate, repeat testing that reveals a persistent elevation in blood lactate may indicate incomplete hemodynamic resuscitation and should prompt efforts, as needed, to further promote hemodynamic stability.

Rationale: Although blood lactate is not a direct measure of tissue perfusion, increased lactate is associated with worse out-comes in children (11). Only one pediatric observational study of lactate-guided resuscitation, which included 77 children with sepsis in the ED, was available (Supplemental Table 9, Supple-mental Digital Content 1, http://links.lww.com/PCC/B138). This study showed that lactate normalization was associated with a decreased risk of persistent organ dysfunction (RR, 0.46; 95% CI, 0.29–0.73 and adjusted RR, 0.47; 95% CI, 0.29–0.78) (56). There is also indirect evidence from adult sepsis, with six RCTs (total of 1,007 patients) evaluating lactate-guided resuscitation of patients with septic shock (216–221). The pooled estimates across all

Page 19: Surviving Sepsis Campaign International Guidelines for the ... · In 2001, the Surviving Sepsis Campaign (SSC) was formed by the Society of Critical Care Medicine (SCCM), European

Copyright © 2020 by the Society of Critical Care Medicine and the World Federation of Pediatric Intensive and Critical Care Societies.Unauthorized reproduction of this article is prohibited

Copyright © 2020 by the Society of Critical Care Medicine and the World Federation of Pediatric Intensive and Critical Care Societies.Unauthorized reproduction of this article is prohibited

Weiss et al

e70 www.pccmjournal.org February 2020 • Volume 21 • Number 2

RCTs showed significant reduction in mortality compared with resuscitation without lactate monitoring (RR, 0.66; 95% CI, 0.55–0.81) (Supplemental Table 9, Supplemental Digital Con-tent 1, http://links.lww.com/PCC/B138). Therefore, although there was not sufficient evidence to propose a recommendation to measure lactate to differentiate low- versus high-risk of sepsis among children with infection or suspected infection (see Rec-ommendation 2), we do suggest that blood lactate levels be used to help guide resuscitation of children with established septic shock or other sepsis-associated organ dysfunction.

VASOACTIVE MEDICATIONS

28) We suggest using epinephrine, rather than dopamine, in children with septic shock (weak recommendation, low quality of evidence).

29) We suggest using norepinephrine, rather than dopamine, in children with septic shock (weak recommendation, very low quality of evidence).

30) We were unable to issue a recommendation for a specific first-line vasoactive infusion for children with septic shock. However, in our practice, we select either epinephrine or norepinephrine as the first-line vasoactive infusion guided by clinician preference, individual patient physiology, and local system factors.

31) We were unable to issue a recommendation about initiat-ing vasoactive agents through peripheral access in children with septic shock. However, in our practice, we often or sometimes administer a dilute concentration of the initial vasoactive medication through a peripheral vein if central venous access is not readily accessible.

Remarks: It is reasonable to begin vasoactive infusions after 40–60 mL/kg of fluid resuscitation if the patient continues to have evidence of abnormal perfusion, or sooner if fluid over-load develops or other concerns for fluid administration are present. Either epinephrine or norepinephrine may be admin-istered through a peripheral vein (or intraosseous, if in place) if central venous access is not readily accessible. Dopamine may be substituted as the first-line vasoactive infusion, admin-istered either peripherally or centrally, if epinephrine or norep-inephrine is not readily available.

Rationale: Epinephrine and norepinephrine both have va-sopressor and inotropic effects, are widely used, and are effective in treating children with fluid-RSS. No studies directly compare epinephrine with norepinephrine. However, epinephrine has

been compared with dopamine in two RCTs in children with fluid-RSS (222, 223). Across both studies, epinephrine was as-sociated with a lower risk of mortality (RR, 0.63; 95% CI, 0.40–0.99) and more organ failure-free days among survivors by day 28 (MD 4 more days; 95% CI, 2.0–6.0) (Supplemental Table 10 and Supplemental Fig. 4, Supplemental Digital Content 1, http://links.lww.com/PCC/B138). Norepinephrine has not been studied in children with septic shock, but in a random-ized trial of norepinephrine versus saline in sedated, mechan-ically ventilated children, mortality was not different between groups (RR, 0.50; 95% CI, 0.10–2.43; Supplemental Table 11a, Supplemental Digital Content 1, http://links.lww.com/PCC/B138) but the norepinephrine group showed higher urine output (p = 0.016) and improved blood pressure (p = 0.04) suggesting improved perfusion relative to saline (224). Ev-idence from adult trials (Supplemental Table 11b, Supple-mental Digital Content 1, http://links.lww.com/PCC/B138) shows a lower mortality rate (RR, 0.93; 95% CI, 0.86–1.00) and lower incidence of arrhythmias (RR, 0.48; 95% CI, 0.40–0.58) with norepinephrine than with dopamine, and no difference in mortality with epinephrine than with norepinephrine (RR, 0.96; 95% CI, 0.77–1.21) (225).

Evidence is insufficient to recommend either epinephrine or norepinephrine as the initial vasoactive agent for children with fluid-RSS. In a survey of our panel members, an equal number used epinephrine and norepinephrine as the first-line vasoactive medication with a general preference for epineph-rine to treat myocardial dysfunction and low cardiac output and for norepinephrine to increase systemic vascular resist-ance. It therefore seems reasonable to use either epinephrine or norepinephrine as the initial vasoactive agent, with the choice made based on individual patient physiology, clinician preference, and local system factors. Once cardiac ultrasound/echocardiography or other advanced monitoring is available, selection of vasoactive therapy should be driven by individual patient physiology.

No pediatric data identify when shock becomes “fluid-refractory” and, thus, to guide when to start vasoactive infu-sions. However, excessive fluid resuscitation can lead to fluid overload, which has been associated with increased mortality in critically ill children (226). A trial comparing a fluid-sparing strategy with early initiation of vasoactive medications com-pared to a fluid-liberal resuscitation strategy is currently on-going (SQUEEZE trial, ClinicalTrials.gov NCT03080038). Until further data are available, we consider it reasonable to begin

TABLE 5. Normal Ranges for Advanced Monitoring

Variable FormulaNormal Range Units

CI CI = cardiac output/body surface area 3.5–5.5 L/min/m2

SI SI = CI/heart rate 30–60 mL/m2

SVRI SVRI = 80 × (mean arterial pressure–central venous pressure)/CI

800–1600 dyne-s/cm5/m2

CI = cardiac index, SI = stroke index, SVRI = systemic vascular resistance index.

Page 20: Surviving Sepsis Campaign International Guidelines for the ... · In 2001, the Surviving Sepsis Campaign (SSC) was formed by the Society of Critical Care Medicine (SCCM), European

Copyright © 2020 by the Society of Critical Care Medicine and the World Federation of Pediatric Intensive and Critical Care Societies.Unauthorized reproduction of this article is prohibited

Online Special Article

Pediatric Critical Care Medicine www.pccmjournal.org e71

vasoactive infusions after 40–60 mL/kg of fluid resuscitation if the patient continues to have evidence of abnormal perfusion. Additional fluid resuscitation may be concurrently adminis-tered if the patient demonstrates physiologic improvement fol-lowing each fluid bolus and without signs of fluid overload.

All vasoactive agents, including norepinephrine, may be ini-tiated through peripheral venous (or intraosseous, if in place) access if central venous access is not readily available to avoid delays in therapy (227, 228). However, central venous access should be obtained as soon as reasonably practicable. In our practice, 82% of panel members reported at least sometimes administering the initial vasoactive infusion peripherally if central venous or intraosseous access was not readily available, particularly in the emergency department or other non-PICU settings. Most panelists preferred epinephrine or dopamine to norepinephrine if peripheral infusion was needed. Although epinephrine or norepinephrine is the preferred first-line med-ication, dopamine may be substituted as the first-line vasoac-tive infusion, administered either peripherally or centrally, if neither epinephrine nor norepinephrine is readily available.

32) We suggest either adding vasopressin or further titrating catecholamines in children with septic shock who require high-dose catecholamines (weak recommendation, low quality of evidence).

Remarks: No consensus was achieved on the optimal threshold for initiating vasopressin. Therefore, this decision should be made according to individual clinician preference.

Rationale: Vasopressin-receptor agonists (vasopressin or terlipressin) have been studied in three RCTs in children (Sup-plemental Table 12, Supplemental Digital Content 1, http://links.lww.com/PCC/B138). Vasopressin was compared with sa-line in one study in children with vasodilatory shock (229) and in one study of children with severe lung disease (230). Terli-pressin was compared with usual care in children with septic shock (231). The mortality rate (RR, 1.14; 95% CI, 0.80–1.62) and ischemic events (RR, 1.56; 95% CI, 0.41–5.91) were higher vasopressin/terlipressin. There were fewer vasoactive-free days with vasopressin (median 25.2 d in vasopressin [interquartile range (IQR) 0.0–28.3], median 27.5 d in control [IQR, 23.1–28.9]). In six RCTs in adults, renal replacement therapy was required less often with vasopressin (RR, 0.74; 95% CI, 0.51–1.08) (232). Weighing the benefit of avoiding renal replace-ment therapy against the potential harm from ischemic events and the nonsignificant difference in mortality, we suggest that vasopressin may be added or catecholamines may be further titrated in children on high doses of catecholamines.

33) We were unable to issue a recommendation about adding an inodilator in children with septic shock and cardiac dys-function despite other vasoactive agents. However, in our practice, we sometimes use inodilators in children with septic shock and evidence of persistent hypoperfusion and cardiac dysfunction despite other vasoactive agents.

Rationale: There are no RCTs of inodilators (including milrinone, dobutamine, or levosimendan) in children with

septic shock with persistent hypoperfusion and cardiac dys-function. A report of two children described improvement in cardiac output with addition of inodilators (233). A case series of 10 children with meningococcal septic shock treated with milrinone described improved core-to-peripheral temperature gradient, with stable blood pressure and no change in acidosis (234). These data were not sufficient to formulate a recom-mendation. However, in our practice, 77% of panel members reported at least sometimes using inodilators in children with septic shock who had evidence of persistent hypoperfusion and cardiac dysfunction despite other vasoactive agents, typically in a PICU with advanced hemodynamic monitoring available.

VENTILATION

34) We were unable to issue a recommendation about whether to intubate children with fluid-refractory, catechol-amine-resistant septic shock. However, in our practice, we commonly intubate children with fluid-refractory, catecholamine-resistant septic shock without respiratory failure.

Rationale: There are no RCTs and/or observational stud-ies of children receiving early intubation for refractory shock without respiratory failure compared with delayed or no intu-bation for the same condition, nor is there suitable indirect ev-idence to substantiate a formal recommendation. However, it is well understood that a high metabolic demand from refrac-tory shock typically indicated by progressive lactic acidemia and end-organ dysfunction can be, at least in part, mitigated by early invasive mechanical ventilation even without clinical symptoms of acute pulmonary edema or respiratory failure (235–237). Furthermore, chest radiograph findings can “lag” behind clinical deterioration (238, 239) such that patients with refractory shock and a “negative” chest radiograph may still progress toward more overt ARDS. Lung ultrasound may pro-vide an alternative tool to chest radiograph in detecting lung pathology, but its utility to identify which sepsis patients may benefit from early mechanical ventilation is not yet clear(240–243). For these reasons, 48% of panel members often or always and 35% sometimes intubate children with fluid-refractory, catecholamine-resistant septic shock even in the absence of clear respiratory failure, while 17% rarely or never do so. Of note, when intubating, caution should be exercised to avoid worsening hypotension or precipitating cardiac arrest as medi-cations used for inducing anesthesia at the time of tracheal in-tubation, along with conversion from spontaneous breathing to use of positive pressure ventilation, may result in a transient deterioration in patient hemodynamics. The panel does rec-ognize that in some settings, invasive mechanical ventilation may not be available or feasible—or may even be detrimental. In these instances, transport of the patient to a higher level of care can be life-saving.

35) We suggest not to use etomidate when intubating children with septic shock or other sepsis-associated organ dysfunc-tion (weak recommendation, low quality of evidence).

Page 21: Surviving Sepsis Campaign International Guidelines for the ... · In 2001, the Surviving Sepsis Campaign (SSC) was formed by the Society of Critical Care Medicine (SCCM), European

Copyright © 2020 by the Society of Critical Care Medicine and the World Federation of Pediatric Intensive and Critical Care Societies.Unauthorized reproduction of this article is prohibited

Copyright © 2020 by the Society of Critical Care Medicine and the World Federation of Pediatric Intensive and Critical Care Societies.Unauthorized reproduction of this article is prohibited

Weiss et al

e72 www.pccmjournal.org February 2020 • Volume 21 • Number 2

Rationale: Etomidate is a short-acting IV anesthetic agent that has been used for inducing anesthesia and sedation for tracheal intubation in patients with unstable hemodynamics. However, concerns regarding the drug’s effect on adrenal func-tion have been raised in adult studies. No RCTs exist in critically ill children with or without sepsis comparing etomidate to an-other anesthesia/sedative regimen. Two observational studies included children. One study from 1984 (244) enrolled acutely injured adults and children (44 intubated with etomidate vs 90 intubated with a benzodiazepine and opioid). A more re-cent study (245) enrolled children with meningococcal sepsis or septic shock with 23 intubated with etomidate as compared with 37 intubated with any other combination of sedatives. Al-though caution must be taken given the small sample size, each of these studies reported higher mortality after use of etomi-date (pooled OR, 4.51; 95% CI, 1.82–11.16) (Supplemental Table 13, Supplemental Digital Content 1, http://links.lww.com/PCC/B138). In addition, den Brinker et al (245) reported a significant association of etomidate with adrenal insuffi-ciency, with cortisol to adrenocorticotropin hormone (ACTH) ratios decreasing by 83% after etomidate exposure. Indirect ev-idence is available from four RCTs in adults (246–249). In the largest of these trials, Jabre et al (249) compared 234 critically ill adults intubated with etomidate to 235 intubated with an al-ternative medication regimen and found higher adrenal insuf-ficiency in the etomidate group (OR, 1.79; 95% CI, 1.37–2.36). Pooled odds of all four adult studies was 1.89 (95% CI, 1.47–2.44) with all studies suggesting significantly increased risk of adrenal insufficiency after etomidate administration. Impor-tantly, this effect was seen even after one dose of etomidate. Unfortunately, there is no conclusive evidence to recommend an optimal alternative induction agent to etomidate, although ketamine and fentanyl are routinely available and can offer fa-vorable hemodynamic profiles in the setting of shock.

36) We suggest a trial of noninvasive mechanical ventilation (over invasive mechanical ventilation) in children with sepsis-induced pediatric ARDS (PARDS) without a clear indication for intubation and who are responding to initial resuscitation (weak recommendation, very low quality of evidence).

Remarks: When noninvasive mechanical ventilation is initi-ated, clinicians should carefully and frequently reevaluate the patient’s condition.

Rationale: Noninvasive mechanical ventilation with con-tinuous positive airway pressure ventilation or bi-level posi-tive airway pressure ventilation may allow for decreased work of breathing and improved oxygenation in the face of sepsis-induced PARDS. Therefore, it is possible to avoid intubation in sepsis patients who are identified early with mild PARDS phys-iology and no evidence of advancing end-organ dysfunction. However, no RCTs in either critically ill children or children with sepsis-induced PARDS compare the effect of noninvasive ventilation to invasive mechanical ventilation on clinical out-comes. Observational studies have tested whether noninvasive mechanical ventilation could mitigate the need for invasive

mechanical ventilation but none specifically focused on chil-dren with sepsis (250–256). We undertook a meta-analysis of three observational studies that evaluated the association of noninvasive mechanical ventilation with mortality in a general PICU population (252, 254, 257). Using unadjusted estimates pooled from the data across all three studies, we found non-invasive ventilation to be associated with a decreased risk of death (RR, 0.21; 95% CI, 0.09–0.47) (Supplemental Fig. 5, Supplemental Digital Content 1, http://links.lww.com/PCC/B138). One additional RCT in immunocompromised chil-dren with acute respiratory dysfunction did not find that early noninvasive ventilation reduced intubation compared with standard care, but the trial was small (42 participants) due to low consent and overall slow recruitment and the direct rele-vance to children with sepsis-induced PARDS without a clear indication for intubation and who are responding to initial resuscitation was not clear (258). Thus, it is reasonable to try noninvasive mechanical ventilation in children with sepsis-induced PARDS who do not have a clear indication for intuba-tion. However, noninvasive ventilation should be reserved for children with sepsis who are responding to initial resuscitation, do not have evidence for ongoing or worsening end-organ dys-function, and in whom close monitoring and frequent reevalu-ation can be ensured (253, 255, 259). This recommendation for children with sepsis-induced PARDS aligns with the 2015 PALICC (260) and 2017 Pediatric Mechanical Ventilation Con-sensus Conference (PEMVECC) (261) guidelines.

37) We suggest using high positive end-expiratory pressure (PEEP) in children with sepsis-induced PARDS (weak rec-ommendation, very low quality of evidence).

Remarks: The exact level of high PEEP has not been tested or determined in PARDS patients. Some RCTs and observa-tional studies in PARDS have used and advocated for use of the ARDS-network PEEP to Fio

2 grid though adverse hemo-

dynamic effects of high PEEP may be more prominent in chil-dren with septic shock.

Rationale: PEEP helps to prevent alveolar collapse, restore end-expiratory lung volume, and improve mean airway pres-sures, all of which help to improve adequate oxygenation in PARDS patients and minimize unnecessary use of high Fio

2.

Adult ARDS patients have been successfully managed with judicious and strict application of a PEEP/Fio

2 grid, initially

implemented in an ARDS-network trial (262). This grid has been applied in children with PARDS enrolled in RCTs (263), but a pediatric-specific PEEP/Fio

2 grid has not been deter-

mined or validated. In 2017, a multicenter observational study by the Collaborative Pediatric Critical Care Research Network reported that pediatric critical care clinicians almost uniformly limit PEEP to 10 cm H

2O irrespective of oxygenation and Fio

2

(264). This is in contrast to the PEMVECC (261) and PALICC (24) recommendations for use of PEEP in excess of 15 cm H

2O

for severe PARDS patients. Our panel reviewed several obser-vational studies of PARDS patients, all published since 2007, each including 12–30% sepsis-induced PARDS (264–276). The largest, a multicenter study by Khemani et al (276), evaluated

Page 22: Surviving Sepsis Campaign International Guidelines for the ... · In 2001, the Surviving Sepsis Campaign (SSC) was formed by the Society of Critical Care Medicine (SCCM), European

Copyright © 2020 by the Society of Critical Care Medicine and the World Federation of Pediatric Intensive and Critical Care Societies.Unauthorized reproduction of this article is prohibited

Online Special Article

Pediatric Critical Care Medicine www.pccmjournal.org e73

1,134 PARDS patients of whom 26% were managed with lower PEEP relative to ARDS Network (ARDSnet) protocol and ex-perienced greater mortality than those managed in accordance with a higher PEEP strategy as recommended by the ARDSnet PEEP/Fio

2 grid (Supplemental Table 14, Supplemental Digital

Content 1, http://links.lww.com/PCC/B138). After adjustment for relevant comorbidities, pediatric patients managed with a PEEP strategy at or above that recommended by the ARDSnet low PEEP/Fio

2 grid had a decreased odds of death compared

with children managed with PEEP lower than that recom-mended by the ARDSnet low PEEP/Fio

2 grid (adjusted OR,

0.50; 95% CI, 0.31–0.81).The panel concluded that PEEP levels greater than 10 cm

H2O may be necessary with progressive hypoxemia, with the

precise amount of “high” PEEP carefully titrated for each in-dividual while attending to the potential adverse hemody-namic effects of increasing intrathoracic pressure in children with septic shock. Therefore, although the optimal approach to setting PEEP has not yet been determined in children with PARDS, carefully increasing PEEP for children with sepsis-induced PARDS who require Fio

2 exceeding 60% and/or ex-

hibit ongoing hypoxemia is reasonable, rather than continuing to manage such children with a low-PEEP or moderate-PEEP strategy of less than or equal to 10 cm H

2O.

38) We cannot suggest for or against the use of recruitment maneuvers in children with sepsis-induced PARDS and re-fractory hypoxemia.

Remarks: If a recruitment maneuver is considered, the use of a stepwise, incremental and decremental PEEP titration ma-neuver is preferred over sustained inflation techniques that have not been optimized through direct testing in PARDS patients. All PARDS patients must be carefully monitored for tolerance of the maneuver.

Rationale: ARDS is characterized by decreased lung com-pliance, risk for atelectasis, and increased intrapulmonary shunt. Recruitment maneuvers have been used in both chil-dren and adults temporarily to increase transpulmonary pres-sure to recruit lung units with the goal of improving both oxygenation and ventilation. Most recruitment maneuvers include either sustained inflation or a step-wise incremental or decremental PEEP titration methodology. However, many clinicians and researchers remain concerned that the optimal strategy for lung recruitment has not been determined and in-judicious implementation of recruitment maneuvers can re-sult in hemodynamic compromise (277), hypercarbia (278), and/or ventilator-induced lung injury (279). PEMVECC did not recommend use of recruitment maneuvers in children, cit-ing an overall lack of evidence in this area (261). In contrast, the 2015 PALICC provided a weak recommendation in favor of recruitment maneuvers with prioritization of a slow stepwise incremental and decremental PEEP method (24).

Two observational studies are potentially informative about use of recruitment maneuvers in children with sepsis-induced PARDS (267, 268). (Supplemental Table 15, Supplemental Digital Content 1, http://links.lww.com/PCC/B138) First,

Boriosi et al (280) enrolled 21 children with lung injury, of whom 66% had sepsis, and used incremental PEEP recruit-ment maneuvers. Patients experienced improved oxygenation as measured by both the Pao

2 to Fio

2 (P/F) ratio and alveolar-

to-arterial oxygen gradient for the 4 hours after recruitment. Second, Duff et al (281) enrolled 32 children and used the sus-tained inflation technique, which also resulted in improved oxygenation for the ensuing 6 hours. However, neither study tested the association of recruitment maneuvers with clinical outcomes, such as ventilator days or mortality. Consequently, despite the potential for benefit for some patients coupled with the possibility of harm (282, 283), insufficient data do not allow us to recommend either for or against recruitment maneuvers in sepsis-induced PARDS patients at this time.

39) We suggest a trial of prone positioning in children with sepsis and severe PARDS (weak recommendation, low quality of evidence).

Remarks: Research trials in adults with ARDS and children with PARDS have emphasized prone positioning for at least 12 hours per day, as tolerated.

Rationale: Prone positioning almost uniformly improves oxygenation in adults with ARDS and children with PARDS. Although the exact mechanisms continue to be elucidated, prone position has been shown to recruit areas of collapsed, de-recruited lung with resultant improved elastance, decreased lung stress and strain, and improved functional residual ca-pacity (284). Given that pulmonary perfusion is thought to be consistent both dorsally and ventrally, an improvement in lung aeration can be met with continued perfusion, thereby re-ducing ventilation-perfusion mismatching (285). Most recent RCTs in adults support use of prone positioning as a poten-tially life-saving management strategy (Supplemental Table 16, Supplemental Digital Content 1, http://links.lww.com/PCC/B138), especially in those meeting severe ARDS criteria (i.e., P/F < 150 mm Hg) (286). This benefit is seen particularly in patients who are positioned for prolonged periods of time, most commonly reported as 12–20 hours per day. Two pediatric RCTs tested the use of prone positioning in PARDS patients (263, 287). Pooled analyses of these two studies yielded a RR of 0.99 (95% CI, 0.36–2.69) for mortality in prone positioning as compared with supine positioning for this patient popula-tion (Supplemental Table 16 and Supplemental Fig. 6, Supple-mental Digital Content 1, http://links.lww.com/PCC/B138). Importantly, no serious adverse events were reported in these trials, although the prone positioning methodology was pro-tocolized in each with particular attention to avoid accidental endotracheal extubation and pressure injury. PALICC (24) did not recommend routine use of prone positioning in PARDS patients but suggested its consideration in severe PARDS. The panel noted that the National Institutes of Health has approved and funded an international RCT of prone positioning in se-vere PARDS (ClinicalTrials.gov identifier NCT02902055).

40) We recommend against the routine use of inhaled nitric oxide (iNO) in all children with sepsis-induced PARDS (strong recommendation, low quality of evidence).

Page 23: Surviving Sepsis Campaign International Guidelines for the ... · In 2001, the Surviving Sepsis Campaign (SSC) was formed by the Society of Critical Care Medicine (SCCM), European

Copyright © 2020 by the Society of Critical Care Medicine and the World Federation of Pediatric Intensive and Critical Care Societies.Unauthorized reproduction of this article is prohibited

Copyright © 2020 by the Society of Critical Care Medicine and the World Federation of Pediatric Intensive and Critical Care Societies.Unauthorized reproduction of this article is prohibited

Weiss et al

e74 www.pccmjournal.org February 2020 • Volume 21 • Number 2

41) We suggest using iNO as a rescue therapy in children with sepsis-induced PARDS and refractory hypoxemia after other oxygenation strategies have been optimized (weak recommendation, moderate quality of evidence).

Rationale: The presumptive mechanism of sepsis-induced PARDS involves alveolar epithelial injury, vascular endothelial injury, and activation of inflammatory, fibrosis, and coagula-tion cascades. As such, PARDS is not a disease process prima-rily of pulmonary arterial hypertension, the therapeutic target of iNO therapy, and so is not recommended for routine use in children with sepsis-associated PARDS. Nonetheless, many PARDS patients have comorbidities that include risk for pul-monary hypertension (e.g., chronic lung disease after prema-turity, congenital heart disease after repair or palliation) or clinical features, such as acidemia and hypoxemia that increase pulmonary arterial pressures. Thus, iNO therapy may be con-sidered in children with documented pulmonary hypertension or severe right ventricular dysfunction (239, 288). Such use of iNO in sepsis must be balanced against its lack of availability or high cost in many areas of the world and, that once in place, iNO use carries a potential patient safety consideration as in-advertent and abrupt discontinuation of the therapy can result in a rapid and potentially life-threatening rebound pulmonary hypertensive crisis.

Several small RCTs (289–291) and observational studies have described significant improvement in oxygenation after iNO therapy (292). Many, but not all, of these studies include patients with sepsis (290, 291, 293–296), and few analyze longer term, clinically relevant outcomes such as mortality. A 2016 Cochrane review indicated no mortality benefit from iNO administration (RR, 0.78; 95% CI, 0.51–1.18) in three RCTs (297). Our analysis of two recent observational stud-ies, one conducted in children on ECMO and another in children with severe PARDS, respectively, suggest possible increased mortality risk (294, 296), whereas one RCT of 55 PARDS patients indicated improved duration of mechanical ventilation in PARDS survivors (291) (Supplemental Table 17 and Supplemental Fig. 7, Supplemental Digital Content 1, http://links.lww.com/PCC/B138). Taken together, these data do not support “routine” use of iNO in all children with sepsis-induced PARDS but do raise the potential for benefit as an emergency rescue therapy for severe, sepsis-induced PARDS with refractory hypoxemia after other oxygenation strategies have been optimized. Emergency rescue use of iNO may allow time to realize benefit from other therapies, such as lung recruitment, or provide a bridge to ECMO or another intervention. However, when iNO is used, we agree with the PALICC recommendation that “assessment of ben-efit must be undertaken promptly and serially to minimize toxicity and to eliminate continued use without established effect” (24). These recommendations align with the 2004 guidelines for use of iNO therapy in neonates and children issued by the European Society for Pediatric and Neonatal Intensive Care (288), PALICC guidelines (24), and a 2017 Cochrane review (292) as no relevant change in evidence has become available.

42) We were unable to issue a recommendation to use high-frequency oscillatory ventilation (HFOV) versus conven-tional ventilation in children with sepsis-induced PARDS. However, in our practice, there is no preference to use or not use HFOV in patients with severe PARDS and refrac-tory hypoxia.

Rationale: HFOV provides a sustained mean airway pres-sure with superimposed high frequency, pendelluft-type, os-cillatory breaths that may improve oxygenation in patients with moderate-to-severe lung disease while minimizing bar-otrauma, volutrauma, and atelectrauma. However, the most efficacious timing of application, optimal settings, and ideal population of patients likely to benefit have not been well es-tablished. HFOV may be difficult to apply effectively in centers with little experience and is not universally available. Despite these practical limitations, both PALICC (24) and PEMVECC (261) endorsed cautionary use of HFOV as an alternative type therapy in patients with severe PARDS. In our panel, clini-cians who use versus those who do not use HFOV in patients with severe PARDS and refractory hypoxia were nearly evenly distributed.

Application of HFOV in adult ARDS patients has yielded concerning results due to a potentially increased mor-tality observed in the adult OSCILLATE RCT (298) and a neutral result in the adult OSCAR RCT (299). Pediatric data include two observational studies with a non-HFOV control group and three randomized trials. In the two ob-servational studies, oxygenation improved with HFOV relative to conventional ventilation but there was a nonsig-nificant trend toward increased mortality (Guo et al [300]: 34.6% vs 22.7%; adjusted OR, 2.74; 95% CI, 0.52–14.6 and Bateman et al [301]: 25% vs 17%; adjusted OR, 1.28; 95% CI, 0.92–1.79). Among three small RCTs, however, a trend toward reduced mortality in those managed with HFOV was observed (pooled RR, 0.77; 95% CI, 0.43–1.36) (302–304). A large, multicenter, international RCT of HFOV compared with conventional mechanical ventilation in severe PARDS patients, including children with and without sepsis, is un-derway and will seek to address many of these issues (www.clinicaltrials.gov/ NCT02902055).

43) We suggest using neuromuscular blockade in children with sepsis and severe PARDS (weak recommendation, very low quality of evidence).

Remarks: The exact duration of neuromuscular blockade to use in severe PARDS patients has not been determined to date. Most of the adult RCT data and pediatric observational data support treatment for 24–48 hours after ARDS onset.

Rationale: Indirect evidence from three adult RCTs (305–307) found that early use of neuromuscular blocking agents (NMBAs) for up to 48 hours in adults with severe ARDS, defined as P/F ratio less than 150 mm Hg, improved 90-day survival and shortened duration of mechanical ventilation without increas-ing muscle weakness. In a multi-center double-blind RCT (307), 340 patients with early severe ARDS, meeting criteria within 48 hours, were randomized to receive either cisatracurium besylate

Page 24: Surviving Sepsis Campaign International Guidelines for the ... · In 2001, the Surviving Sepsis Campaign (SSC) was formed by the Society of Critical Care Medicine (SCCM), European

Copyright © 2020 by the Society of Critical Care Medicine and the World Federation of Pediatric Intensive and Critical Care Societies.Unauthorized reproduction of this article is prohibited

Online Special Article

Pediatric Critical Care Medicine www.pccmjournal.org e75

or placebo once adequately sedated. After adjustment for base-line P/F, plateau pressure, and the Simplified Acute Physiology Score, the cisatracurium group had a hazard ratio for death at 90 days of 0.68 (95% CI, 0.48–0.98) compared with the placebo group. Early use of NMBAs was also associated with decreased organ system dysfunction, less air leak, and a decreased pro-inflammatory response (308). These findings remained con-sistent when combined with earlier smaller studies from the same group of investigators in a meta-analysis. However, the more recent ROSE trial of early neuromuscular blockade in adults with moderate to severe ARDS was stopped futility at the second interim analysis (enrollment of 1,006 patients) with a 90-day mortality difference of 42.5% in the intervention group versus 42.8% in the control group. In this study, the in-tervention group received continuous cisatracurium and deep sedation for 48 hours compared with the control group that re-ceived lighter sedation targets (Richmond Agitation Scale of 0 to –1). Both groups received low tidal volume ventilation with high PEEP strategy. Notably, only 13.8% of patients enrolled in ROSE had nonpulmonary sepsis as a primary diagnosis.

In pediatrics, there are no prospective data regarding the use of NMBAs in PARDS (with or without sepsis), al-though there is an ongoing pediatric trial in the Netherlands (ClinicalTrials.gov NCT02902055). In one large retrospective study of 317 children with PARDS, of whom 23% experienced sepsis-induced PARDS (309), mortality was lower in those chil-dren treated with neuromuscular blockade (8.8% vs 17.7%). However, duration of mechanical ventilation was longer in the treatment group and proportion with neuromuscular weak-ness was not assessed (Supplemental Table 18, Supplemental Digital Content 1, http://links.lww.com/PCC/B138).

CORTICOSTEROIDS

44) We suggest against using IV hydrocortisone to treat chil-dren with septic shock if fluid resuscitation and vaso-pressor therapy are able to restore hemodynamic stability (weak recommendation, low quality of evidence).

45) We suggest that either IV hydrocortisone or no hydro-cortisone may be used if adequate fluid resuscitation and vasopressor therapy are not able to restore hemodynamic stability (weak recommendation, low quality of evidence).

Rationale: A potential role for IV hydrocortisone as ad-junctive therapy for septic shock is supported by various roles of cortisol in homeostasis and the stress response. For example, cortisol directly decreases reuptake of norepinephrine (310), and enhances calcium availability in myocardial and vascular smooth muscle cells (311) promoting myocardial contractility and vasoconstriction, respectively. Cortisol helps to inhibit prostacyclin and endogenous nitric oxide production, resulting in increased vascular tone (312), modulation of capillary leak (313), and augmentation of the beta-adrenergic receptor in the heart (312). However, potential adverse side effects of corti-costeroid therapy include hyperglycemia (314, 315), catabo-lism-related diffuse neuromuscular weakness (including the diaphragm) (316, 317), and hospital-acquired infections (318).

These effects may be under-appreciated in critically ill patients but can contribute to worse outcomes (319).

At least one pediatric (320) and several adult (321) inter-ventional trials examining adjunctive corticosteroids for septic shock have concluded that this drug class hastens resolution of shock. Of the four adult high-quality contemporary RCTs, two reported a mortality reduction and two did not (322–326). A recent meta-analysis of 42 RCTs including 9,969 adults and 225 children with sepsis found that corticosteroids possibly result in a small reduction in short-term mortality (RR, 0.93; 95% CI, 0.84–1.03), long-term mortality (0.94; 95% CI, 0.89–1.00), faster resolution of shock, and shorter LOS, while also possibly increasing the risk of neuromuscular weakness (RR, 1.21; 95% CI, 1.01–1.52) (327). Despite a weak recommenda-tion to treat sepsis with hydrocortisone based on the findings noted in the overall meta-analysis (328), the pediatric studies enrolled a combined small number of subjects, reported in-consistent conclusions, had methodologic limitations, and did not demonstrate an overall mortality reduction (320, 329–331) (Supplemental Table 19, Supplemental Digital Content 1, http://links.lww.com/PCC/B138).

Observational cohort studies have reported either harm or no benefit with hydrocortisone in children with septic shock (5, 332–336). For example, a retrospective analysis of the REsearching severe Sepsis and Organ dysfunction in children: a gLobal perspectiVE trial of activated protein C in pediatric sepsis found no differences in mortality, duration of mechan-ical ventilation and vasoactive-inotropic support, or PICU stay among 193 children who received and 284 who did not receive open-labeled corticosteroids (333). Despite the post hoc analysis, age, sex, Pediatric Risk of Mortality (PRISM) III scores, baseline number of dysfunctional organs, and baseline Pediatric Overall Performance Category scores did not differ between corticosteroid-treated and corticosteroid nontreated groups.

Several pediatric and adult studies have attempted to use random cortisol and/or cosyntropin-stimulated cortisol serum concentrations to identify which patients with septic shock may benefit from hydrocortisone therapy, but reliable cutoffs have not been clearly identified. Challenges relate to variability in 1) the cortisol assay itself; 2) cortisol metabo-lism (11-beta-hydroxysteroid dehydrogenase) during sepsis; 3) corticosteroid-binding globulin concentrations; and 4) mul-tiple tissue (e.g., elastase, anti-glucocorticoid compounds) and cellular (e.g., glucocorticoid receptor) factors. Therefore, use of random cortisol or stimulation tests to guide corticosteroid prescription in children with septic shock cannot be recom-mended as this time. However, for any patient with a clinical concern for primary adrenal insufficiency (e.g., a patient with significant and unexplained hypoglycemia, hyponatremia, and/or hyperkalemia), a high-dose cosyntropin-stimulation test should be performed. Interpretation should focus on the baseline serum ACTH concentration (above normal indicating primary adrenal insufficiency) and the 60-minute stimulated serum cortisol concentration (< 18 µg/dL indicating primary adrenal insufficiency) (337).

Page 25: Surviving Sepsis Campaign International Guidelines for the ... · In 2001, the Surviving Sepsis Campaign (SSC) was formed by the Society of Critical Care Medicine (SCCM), European

Copyright © 2020 by the Society of Critical Care Medicine and the World Federation of Pediatric Intensive and Critical Care Societies.Unauthorized reproduction of this article is prohibited

Copyright © 2020 by the Society of Critical Care Medicine and the World Federation of Pediatric Intensive and Critical Care Societies.Unauthorized reproduction of this article is prohibited

Weiss et al

e76 www.pccmjournal.org February 2020 • Volume 21 • Number 2

In summary, no high-quality investigations currently sup-port or refute the routine use of adjunctive corticosteroids for pediatric septic shock or other sepsis-associated organ dys-function. At the time of this publication, an RCT is in prog-ress to examine the potential risks and benefits of adjunctive hydrocortisone for fluid and vasoactive-inotropic recalcitrant septic shock in children. However, this uncertainty does not apply to children presenting with septic shock or other sepsis-associated organ dysfunction who also have acute or chronic corticosteroid exposure, hypothalamic-pituitary-adrenal axis disorders, congenital adrenal hyperplasia or other corticoste-roid-related endocrinopathies, or have recently been treated with ketoconazole or etomidate, for whom prescription of stress-dose hydrocortisone is indicated, with or without evalu-ation of the adrenal axis (338).

ENDOCRINE AND METABOLIC

46) We recommend against insulin therapy to maintain a blood glucose target at or below 140 mg/dL (7.8 mmol/L) (strong recommendation, moderate quality of evidence).

47) We were unable to issue a recommendation regarding what blood glucose range to target for children with septic shock or other sepsis-associated organ dysfunction. However, in our practice, there was consensus to target blood glucose levels below 180 mg/dL (10 mmol/L) but there was not consensus about the lower limit of the target range.

Rationale: Although hyperglycemia has been associated with poor outcomes in numerous studies of critically ill chil-dren and adults, three prospective multicenter RCTs of glucose control to a low target range (including 50–80, 70–100, 72–126, 80–110 mg/dL or 2.8–4.4, 3.9–5.6, 4.0–7.0, 4.4–6.1 mmol/L) have not demonstrated clinical benefit in children (339–341) (Supplemental Table 20, Supplemental Digital Content 1, http://links.lww.com/PCC/B138). One single-center RCT did show substantial mortality benefit, but there was a high rate of severe hypoglycemia and the higher target range cohort had substantially higher blood glucose levels than those used in the other multicenter RCTs (342). A trial involving children with burn injuries, a unique PICU population, demonstrated no mortality benefit but did find a significant reduction in mor-bidity (343). Notably, all trials included sepsis patients but none targeted them exclusively. Meta-analyses of all published prospective trials in children have shown no clinical benefits overall, but showed a substantially higher risk of hypoglycemia when using insulin therapy to maintain a glucose target below 140 mg/dL (7.8 mmol/L) (344, 345). Even brief episodes of se-vere hypoglycemia during septic shock in children may be a risk factor for poor long-term developmental outcomes (346–349).

Treating hyperglycemia greater than or equal to 180 mg/dL (≥ 10 mmol/L) may be desirable as incidence of insulin-induced hypoglycemia in the studied pediatric cohorts with targets of 140–180 mg/dL (7.8–10.0 mmol/L) is extremely low. There are, however, no direct comparisons between treatment to less than 180 mg/dL (10.0 mmol/L) and no treatment. Therefore, evidence cannot definitively guide this therapeutic target.

However, given that the guidelines for adults recommend an upper limit of 180 mg/dL (10 mmol/L) and given the lack of harm demonstrated in the pediatric trials with those targets, treating children with septic shock or other sepsis-associated organ dysfunction with IV insulin with a goal upper blood glu-cose target of 180 mg/dL (10 mmol/L) is reasonable. The lower target, that is, the glucose concentration below which insulin infusion should be discontinued, has also not been specifi-cally studied, but is reasonable to set at 140–150 mg/dL (7.8–8.3 mmol/L), based on similar principles. In a survey of our panel members, 32.5% always or often and 17.5% sometimes target glucose levels between 140 and 180 mg/dL. Regardless of the glucose target, the overriding goal during insulin therapy should be avoidance of hypoglycemia.

48) We were unable to issue a recommendation as to whether to target normal blood calcium levels in children with septic shock or sepsis-associated organ dysfunction. However, in our practice, we often target normal calcium levels for chil-dren with septic shock requiring vasoactive infusion support.

Rationale: Calcium has an essential role in nearly all cellular processes, including myocardial contractility and vasomotor tone. As such, intracellular and circulating levels of calcium are tightly regulated. During septic shock, derangements in calcium regulation frequently occur in critically ill adults and children. However, a systematic review of adult literature found no evidence to support treating hypocalcemia of critical illness (350). Calcium supplementation may actually worsen organ dysfunction and is correlated with adverse outcomes in critically ill adult patients receiving PN (351). Although the prevalence of hypocalcemia in critically ill children has been reported to be up to 75% and is associated with organ dys-function (352), no studies in children with septic shock have investigated the effect of calcium supplementation to treat hy-pocalcemia. However, in our practice, 65% of panel members always or often and 20% sometimes target normal calcium levels with parenteral calcium administration in children with septic shock requiring vasoactive infusion support. Only 15% of panel members rarely or never target normal calcium levels.

49) We suggest against the routine use of levothyroxine in chil-dren with septic shock and other sepsis-associated organ dysfunction in a sick euthyroid state (weak recommenda-tion, low quality of evidence).

Rationale: Critically ill children, similar to adults, develop low tri-iodothyronine (T3) and low normal thyroxine (T4) concentrations without the compensatory rise in thyroid-stim-ulating hormone that is typical of the “sick euthyroid” state or hypothyroxinemia of nonthyroidal illness (353). The decrease in T3 is due both to increased thyroid hormone turnover and to decreased de-iodination of T4 to T3, with redirection of T4 metabolism toward higher levels of biologically inactive re-verse T3. The magnitude of the drop in T3 within the first 24 hours of illness reflects the severity of illness (354). Although of theoretical benefit, few trials of thyroid hormone replace-ment have been conducted in critically ill children and none in

Page 26: Surviving Sepsis Campaign International Guidelines for the ... · In 2001, the Surviving Sepsis Campaign (SSC) was formed by the Society of Critical Care Medicine (SCCM), European

Copyright © 2020 by the Society of Critical Care Medicine and the World Federation of Pediatric Intensive and Critical Care Societies.Unauthorized reproduction of this article is prohibited

Online Special Article

Pediatric Critical Care Medicine www.pccmjournal.org e77

children with sepsis. Two prospective RCTs in children under-going cardiac surgery (without sepsis) showed no difference in mortality, vasoactive days, or PICU LOS (355, 356). One open-label study in premature neonates also showed no difference in clinical outcomes (357). Taken together, there are no direct data to inform a recommendation for children with sepsis, and no indirect data from other critically ill children to support a recommendation for the routine use of levothyroxine in chil-dren with septic shock and other sepsis-associated organ dys-function in a sick euthyroid state.

50) We suggest either antipyretic therapy or a permissive approach to fever in children with septic shock or other sepsis-associated organ dysfunction (weak recommenda-tion, moderate quality of evidence).

Rationale: Fever is a complex physiologic response associ-ated with sepsis, and it remains unclear whether fever is a ben-eficial (358) or a harmful (359) response to infection. Potential benefits include inhibiting the growth of some pathogens and increased neutrophil production and lymphocyte prolifera-tion. Conversely, fever is associated with an increased metabolic rate (which may or may not have detrimental effects in patients with sepsis) and may impair some components of immune function. Fever can also make patients uncomfortable (360). Thus, the putative benefits of maintaining normothermia by treating fever are unclear.

No direct evidence for or against the use of antipyretics in febrile children with sepsis-associated organ dysfunction exists. Rather, the panel had to consider indirect data extrapo-lated from studies in adults. One systematic review of adult patients studied the use of antipyretics and physical cooling methods included eight RCTs (1,507 patients) and eight obser-vational studies (17,432 patients) (361). This study had 28-day mortality as the primary outcome, with additional outcomes of early mortality (i.e., death on or prior to day 14), frequency of acquisition of hospital-acquired infection, frequency of shock reversal, and mean changes in body temperature, heart rate, and minute ventilation. No difference was noted in 28-day mortality. Effects on early mortality differed between the ran-domized (favored reduced mortality with antipyretic therapy) and observational (favored increased mortality with antipy-retic therapy) studies. Although antipyretic therapy success-fully decreased body temperature, there was no effect on heart rate, minute ventilation, shock reversal, or acquisition of noso-comial infections. This study did not assess outcome measures of patient comfort. Based on available data, we are not able to recommend the optimal approach to fever in children with sepsis. However, it is reasonable to provide antipyretic therapy to optimize patient comfort, to reduce metabolic demand under certain clinical scenarios (e.g., refractory shock, pulmo-nary hypertension), and to reduce extreme body temperatures.

NUTRITION

51) We were unable to issue a recommendation regarding early hypocaloric/trophic enteral feeding followed by slow

increase to full enteral feeding versus early full enteral feeding in children with septic shock or sepsis-associated organ dysfunction without contraindications to enteral feeding. However, in our practice, there is a preference to commence early enteral nutrition within 48 hours of ad-mission in children with septic shock or sepsis-associated organ dysfunction who have no contraindications to en-teral nutrition and to increase enteral nutrition in a step-wise fashion until nutritional goals are met.

Rationale: No studies examine the enteral nutrition ad-vancement strategy in children with septic shock or other sepsis-associated organ dysfunction. Indirect evidence from a small RCT in critically ill children examines early (6–24 hr) versus late enteral nutrition (> 24 hr) in, respectively, 57 and 52 children (362). Early enteral feeding had no effect on du-ration of PICU stay, but a trend toward lower mortality in the early feeding group (30% in early feeding vs 48% in late feeding; p = 0.07) was shown. There is also indirect evidence from the EDEN trial in adults (363) in which 200 patients were randomized to receive either trophic or full enteral feed-ing for the first 6 days. This study demonstrated no differ-ence in number of ventilator-free days, mortality at 60 days, or infectious complications, but trophic enteral feeding was associated with less gastrointestinal intolerance. Because neither of these studies was conclusive nor directly studied children with septic shock or other sepsis-associated organ dysfunction, no evidence-based recommendation could be made by the panel. However, in critically ill children, a step-wise approach to increasing enteral feeds has been shown to reduce time needed to reach nutritional goals (364–367). In our practice, 60% of panel members always or often and 20% sometimes commence early enteral feeding within 48 hours of admission in children with septic shock or sepsis-associ-ated organ dysfunction who have no contraindications to en-teral nutrition, while 20% of panel members rarely or never pursue this practice.

52) We suggest not withholding enteral feeding solely on the basis of vasoactive-inotropic medication administration (weak recommendation, low quality of evidence).

Remarks: Enteral feeding is not contraindicated in children with septic shock after adequate hemodynamic resuscitation who no longer require escalating doses of vasoactive agents or in whom weaning of vasoactive agents has started.

Rationale: We reviewed indirect evidence from three ob-servational studies (two retrospective and one prospective) in postoperative/cardiac pediatric populations. These studies re-ported that enteral feeding was tolerated in patients on nones-calating/weaning doses of vasoactive agents without increased adverse effects or gastrointestinal complications (368–370). In another study of 339 critically ill children, there was no asso-ciation between enteral feeding and the development of severe gastrointestinal outcomes such as vomiting, diarrhea, abdom-inal distension, bleeding, necrotizing enterocolitis, or perfora-tion (368). However, in the report, the decision to start enteral nutrition may have been biased by the clinical condition of

Page 27: Surviving Sepsis Campaign International Guidelines for the ... · In 2001, the Surviving Sepsis Campaign (SSC) was formed by the Society of Critical Care Medicine (SCCM), European

Copyright © 2020 by the Society of Critical Care Medicine and the World Federation of Pediatric Intensive and Critical Care Societies.Unauthorized reproduction of this article is prohibited

Copyright © 2020 by the Society of Critical Care Medicine and the World Federation of Pediatric Intensive and Critical Care Societies.Unauthorized reproduction of this article is prohibited

Weiss et al

e78 www.pccmjournal.org February 2020 • Volume 21 • Number 2

the patient. In a retrospective study of 52 critically ill children, the use of vasoactive medications was not associated with an increase in feeding intolerance or gastrointestinal complica-tions (369). In a prospective observational study of critically ill children who received postpyloric feeding, 44 of 65 patients (67.7%) with shock and 284 of 461 patients (61.6%) without shock received enteral nutrition within 48 hours. Although gastrointestinal complications were more common in chil-dren admitted with shock, no association between the inci-dence of digestive tract complications and early (first 48 hr) or late administration of postpyloric enteral nutrition was re-ported (370). Based on these studies which, while providing indirect evidence, all consistently found that enteral feeding was not associated with harm, we recommend not to withhold enteral nutrition solely because vasoactive-inotropic medica-tions are being used. Current evidence supports starting en-teral nutrition in hemodynamically stable patients who are no longer requiring fluid resuscitation or escalating doses of vasoactive agents.

53) We suggest enteral nutrition as the preferred method of feeding and that parenteral nutrition may be withheld in the first 7 days of PICU admission in children with septic shock or other sepsis-associated organ dysfunction (weak recommendation, moderate quality of evidence).

Rationale: No studies have been published on this spe-cific issue of nutrition in children with septic shock or other sepsis-associated organ dysfunction. However, in a general co-hort of 1,440 critically ill children enrolled in the international multicenter RCT of pediatric early versus late PN in critical illness (371), withholding parenteral nutrition during the first week in PICU when enteral nutrition was less than 80% of pre-scribed goal was clinically superior to providing supplemental parental nutrition within 24 hours of admission (372). Sec-ondary analyses of the PEPaNIC trial showed that withholding PN was also beneficial in term neonates and children who were undernourished at admission (373, 374), although withhold-ing parenteral nutrition in term neonates was also associated with increased risk of severe hypoglycemia (373). A long-term follow-up 2 years after PICU admission showed that with-holding parenteral nutrition for 1 week did not affect survival, anthropometrics, or health status, but did improve certain domains of neurocognitive development (375). Although the results of the PEPaNIC trial corroborated the findings from adult RCTs, the optimal timing of parenteral nutrition in the critically ill child with sepsis is still not clear (371, 376–378). Our recommendation is based on one trial, and therefore, the evidence to withhold PN in the first 7 days of PICU admission is of moderate certainty and must be explored further using pragmatic timing for PN in the first week, particularly in se-verely malnourished patients and neonates.

54) We suggest against supplementation with specialized lipid emulsions in children with septic shock or other sepsis-associated organ dysfunction (weak recommendation, very low quality of evidence).

Rationale: In two RCTs evaluating immunomodulatory formulas, including lipid emulsions, in critically ill children, outcomes were not significantly different (379, 380). One RCT was terminated during interim analysis because of un-likely benefit in the intervention arm (380). In another small RCT, use of enteral feeding supplemented with or without omega-3 fatty acids in 120 critically ill children with sepsis was investigated (381). Univariate analyses showed a signifi-cant difference in inflammatory mediators and reduction in PICU LOS, but these outcome benefits were not evident in the multivariable analyses. Taken together, although prom-ising, insufficient evidence is available to support routine supplementation in pediatric sepsis with specialized lipid emulsions.

55) We suggest against the routine measurements of gastric residual volumes (GRVs) in children with septic shock or other sepsis-associated organ dysfunction (weak recom-mendation, low quality of evidence).

Rationale: Although routine measurement of GRV is a rel-atively common practice in PICUs, there is no direct evidence in pediatric sepsis. In a two-center observational cohort study of critically ill children admitted with a variety of diagnoses, one center reported routine use of GRV monitoring while the other center did not practice GRV measurements (382). The center that advanced enteral nutrition without routine mea-surements of GRV did not have an increase in the incidence of vomiting, ventilator acquired pneumonia, or necrotizing en-terocolitis in comparison with the other PICU (Supplemental Table 21, Supplemental Digital Content 1, http://links.lww.com/PCC/B138). Although there are likely some children for whom measuring GRV would likely be useful (e.g., gastropa-resis, omphalocele, gastroschisis), no evidence supports rou-tine measurements in all patients at this time and, if measured, GRV is not sufficient to diagnose EN intolerance.

56) We suggest administering enteral feeds through a gastric tube, rather than a postpyloric feeding tube, to children with septic shock or other sepsis-associated organ dys-function who have no contraindications to enteral feeding (weak recommendation, low quality of evidence).

Rationale: In three small RCTs, gastric versus postpyloric enteral feeding were compared in mechanically-ventilated children with a variety of diagnoses (383–385). The outcomes reported included lower caloric achievement with gastric feeding and delayed start of enteral feeding with postpyloric feeding (383, 384). No significant difference was found in the incidence of ventilator-associated pneumonia between gastric and postpyloric feeding (385). On the basis of these studies, there is no clear evidence that postpyloric feeding is benefi-cial and there is concern for potential harm through delayed optimization of enteral nutrition. Therefore, we suggest that feeding with a gastric tube is physiologic and, based on current evidence, the preferred method for enteral nutrition. Postpy-loric feeding may be considered in patients in whom gastric feeding is either contraindicated (e.g., high-risk for aspiration)

Page 28: Surviving Sepsis Campaign International Guidelines for the ... · In 2001, the Surviving Sepsis Campaign (SSC) was formed by the Society of Critical Care Medicine (SCCM), European

Copyright © 2020 by the Society of Critical Care Medicine and the World Federation of Pediatric Intensive and Critical Care Societies.Unauthorized reproduction of this article is prohibited

Online Special Article

Pediatric Critical Care Medicine www.pccmjournal.org e79

or was not tolerated/advanced, and as a result, nutritional goals were unable to be met.

57) We suggest against the routine use of prokinetic agents for the treatment of feeding intolerance in children with septic shock or other sepsis-associated organ dysfunction (weak recommendation, low quality of evidence).

Rationale: Prokinetic agents, such as metoclopramide and erythromycin, are often used in the PICU in an effort to reduce feeding intolerance (386). Indirect evidence for this question was provided from the only pediatric randomized control trial, which was a combined intervention of enteral zinc, selenium, glutamine, and IV metoclopramide. In critically ill children, this combined intervention failed to reduce the development of sepsis or incidence of hospital-acquired infection in immuno-competent children, although the intervention including meto-clopramide did reduce the rate of hospital-acquired infection and sepsis in immunocompromised children. However, the ap-plication of this study to children who already have sepsis is not clear. Prokinetic agents are also not without risk as they have been associated with prolongation of the QT interval and ven-tricular arrhythmias (387–389). Further investigation is needed to determine if prokinetic agents are beneficial in patients with sepsis, particularly in immunocompromised children.

58) We suggest against the use of selenium in children with septic shock or other sepsis-associated organ dysfunction (weak recommendation, low quality of evidence).

Rationale: Although clinical research examining the use of selenium among critically ill neonates and adults has been done (Supplemental Table 22, Supplemental Digital Content 1, http://links.lww.com/PCC/B138), there no data regarding selenium supplementation as potential adjunctive therapy for pediatric sepsis. Selenium plays a key role as a cofactor for glutathione peroxidase, iodothyronine deiodinase, and thiore-doxin (390); accordingly, selenium deficiency could affect thy-roid metabolism and the response to oxidative stress during critical illness. Furthermore, low serum selenium concentra-tions are common in critical illness (391, 392) and infection (393), and have been associated with measures of oxidative stress in neonates (394) and adults (395).

A systematic review of investigations examining selenium supplementation in preterm neonates reported improved outcomes, including reduction in occurrence of sepsis (396). Similarly, a published systematic review and meta-analysis of the effect of parenteral selenium supplementation in criti-cally ill adult sepsis patients concluded that this intervention reduced risk of mortality (397), but when the meta-analysis was updated to include the results of a more recent RCT, there was no difference in mortality in those treated with or without selenium supplementation (50). In an interventional trial examining the potential benefit of zinc, selenium, gluta-mine, and metoclopramide administration to critically ill chil-dren, there was no reduction in the primary outcome measure, namely, time until the first episode of nosocomial infection/sepsis (380). Based on lack of interventional trials examining

selenium supplementation in the setting of pediatric sepsis and sepsis-associated organ dysfunction, we suggest against its use as a weak recommendation.

59) We suggest against the use of glutamine supplementation in children with septic shock or other sepsis-associated organ dysfunction (weak recommendation, low quality of evidence).

Rationale: During catabolic stress, the human body is unable to produce adequate quantities of glutamine and, therefore, its essential role as a fuel source for enterocytes and immune cells is diminished. Over the past 2 decades, several investigations of glutamine administration alone and in various combinations with other nutritional supplements have been conducted in critically ill populations (380, 398–404), including those with sepsis (399, 405–407). Contemporary studies have not found glutamine in any form (enteral or parenteral) and/or in combi-nation with other nutritional elements to significantly improve morbidity or mortality in critically ill infants, children, and adults, including those with sepsis (408–410) (Supplemental Table 23, Supplemental Digital Content 1, http://links.lww.com/PCC33/B138). However, single-element studies adminis-tering only glutamine to children with sepsis and septic shock are scarce. An RCT by Jordan et al (401) randomized children (49 control; 49 interventional) with sepsis and septic shock requiring PN to either standard PN or PN with glutamine sup-plementation for the purpose of examining oxidative stress and inflammatory response. This investigation supports earlier studies in broader populations finding no differences in PICU (p = 0.062) or hospital LOS (p = 0.09) or hospital mortality (p = 0.31). Two other studies of glutamine administration in combination with other elements to children with septic shock and critical illness are available (380, 399). The RCT by Brias-soulis et al (399) examined children with septic shock receiving glutamine in combination with arginine, antioxidants, and omega-3 fatty acids. Although the main outcome of change in cytokines showed some promise, no difference was noted between groups for hospital survival (80% vs 87%) or LOS (10.4 ± 2.2 vs 11.4 ± 2.5 d) (15). Carcillo et al (380) random-ized 283 subjects from eight PICUs to a control group receiving whey protein formula or an intervention group receiving for-mula with zinc, selenium, glutamine, and IV metoclopramide supplementation. There was no difference between hospital-acquired infections and clinical sepsis per 100 days (p = 0.81), PICU LOS (p = 0.16), or 28-day mortality (8/139 [5.8%] vs 15/145 [10.3%]). Subjects from this trial were also categorized by immune status with the suggestion that immune status may play a role in the effectiveness of nutritional supplemental, including glutamine (411). However, no direct evidence re-garding glutamine supplementation in children with sepsis exists; hence, we suggest against the use of glutamine therapy in children with septic shock or other sepsis-associated organ dysfunction until further data become available.

60) We suggest against the use of arginine in the treatment of children with septic shock or other sepsis-associated organ

Page 29: Surviving Sepsis Campaign International Guidelines for the ... · In 2001, the Surviving Sepsis Campaign (SSC) was formed by the Society of Critical Care Medicine (SCCM), European

Copyright © 2020 by the Society of Critical Care Medicine and the World Federation of Pediatric Intensive and Critical Care Societies.Unauthorized reproduction of this article is prohibited

Copyright © 2020 by the Society of Critical Care Medicine and the World Federation of Pediatric Intensive and Critical Care Societies.Unauthorized reproduction of this article is prohibited

Weiss et al

e80 www.pccmjournal.org February 2020 • Volume 21 • Number 2

dysfunction (weak recommendation, very low quality of evidence).

Rationale: Reduced availability of arginine in sepsis may lead to decreased endogenous nitric oxide synthesis, loss of microcirculatory regulation, and altered immune response (412–414). In the only pediatric RCT of arginine supplemen-tation in children with sepsis (415), 10 children received infu-sions of arginine and had enhanced arginine oxidation and increased nitric oxide levels, but no clinical outcomes were re-ported. In indirect data from adult studies, RCTs of L-arginine supplementation have been small and have reported both pos-itive and negative effects on mortality (416–420). One trial in septic adults found decreased mortality(418), but other studies found no benefit or increased mortality in adults with sepsis (416, 419, 420). Some authors found improvement in sec-ondary outcomes in patients with sepsis, such as reduced in-fectious complications and shorter LOS, but the relevance of these findings and their applicability to children with sepsis in the face of potential harm is unclear. Hence, in the absence of evidence of demonstrated benefit, we suggest against the use of arginine therapy in children with sepsis-associated organ dys-function until further data become available.

61) We suggest against using zinc supplementation in children with septic shock and other sepsis-associated organ dysfunc-tion (weak recommendation, very low quality of evidence).

Rationale: Alterations in zinc homeostasis and associations between zinc levels and outcomes have been reported in the critically ill. Benefits of zinc supplementation have been shown in some forms of infectious illnesses. However, no trials of zinc supplementation in children with sepsis have been conducted. One RCT in critically ill children comparing daily supplemen-tation with zinc, selenium, glutamine, and metoclopramide versus whey protein was stopped during interim analysis due to futility (380). Based on conflicting studies in the adult liter-ature, routine supplementation of zinc is not recommended in nutritional guidelines for critically ill adults (421). Future RCTs examining the optimal timing and dose of zinc in chil-dren with sepsis and septic shock and its impact on immune re-sponse and clinical outcomes might help answer this question.

62) We suggest against the use of ascorbic acid (vitamin C) in the treatment of children with septic shock or other sepsis-associated organ dysfunction (weak recommendation, very low quality of evidence).

Rationale: Ascorbic acid (vitamin C) has multiple physiologic functions. Most importantly in the setting of sepsis, vitamin C is an antioxidant and neutralizes reactive oxygen and nitrogen radi-cals, inhibits activation of pro-inflammatory cytokines, increases endogenous vasopressor synthesis, and inhibits bacterial replica-tion (422–424). Adults with sepsis frequently have very low lev-els of vitamin C. In one study, 88% of adults with septic shock had hypovitaminosis C (425). Small studies in adults suggest that treatment of septic patients with vitamin C may improve organ dysfunction (426) and reduce mortality (427). Vitamin C has also been used as a component of combination therapy, typically

with thiamine and corticosteroids, in adults with sepsis (428). One study compared such treatment in 47 adult patients with sepsis to historical control patients (429). Treatment was associ-ated with decreased hospital mortality (OR, 0.13; 95% CI, 0.04–0.48), shorter duration of vasopressor therapy, and improved organ dysfunction scores (Supplemental Table 24, Supplemental Digital Content 1, http://links.lww.com/PCC/B138).

Currently, there are no data on the use of vitamin C in crit-ically ill children or in pediatric sepsis. The prevalence of low vitamin C levels in septic children is unknown, and no studies have investigated the effect of vitamin C supplementation, ei-ther alone or in combination with other agents, in the treat-ment of pediatric sepsis.

63) We suggest against the use of thiamine to treat children with sepsis-associated organ dysfunction (weak recom-mendation, low quality of evidence).

Rationale: Thiamine is a crucial factor in cellular metabo-lism. In its active form, thiamine pyrophosphate is an essential coenzyme used to generate energy (adenosine triphosphate) from glucose. The human body does not produce thiamine and, with a short half-life and small body stores, thiamine defi-ciency can develop within days of critical illness and inadequate nutrition, resulting in impaired oxidative and carbohydrate metabolism. Low blood concentrations of thiamine have been reported at admission of critically ill children and adults with sepsis and septic shock (430–432). A study examining thi-amine deficiency in children admitted to the PICU showed that low blood thiamine concentration in those with severe sepsis or septic shock was associated with mortality (OR, 8.40; 95% CI, 1.38–51.0) (431). In an RCT of 88 adults with septic shock (Supplemental Table 25, Supplemental Digital Content 1, http://links.lww.com/PCC/B138), there were no differences between treatment with thiamine versus placebo for the pri-mary outcome of change in lactate levels or the secondary out-comes of mortality, shock reversal, and LOS (430). However, on post hoc analysis, thiamine treatment in the subgroup with thiamine deficiency at admission was associated with lower lactate level within 24 hours and lower mortality (p = 0.047). However, more evidence is needed to recommend whether thi-amine supplementation should be used to treat children with septic shock or other sepsis-associated organ dysfunction. Also, it may be important for this evidence to be considered in the context of thiamine status at PICU admission.

64) We suggest against the acute repletion of vitamin D defi-ciency (VDD) for treatment of septic shock or other sep-sis-associated organ dysfunction (weak recommendation, very low quality of evidence).

Rationale: A systematic review and meta-analysis of 17 studies including 2,783 patients showed that approximately half of critically ill children have VDD (25-hydroxy vitamin D level < 50 nmol/L or < 20 ng/mL) at PICU admission (189). Further, VDD was associated with higher illness severity, mul-tiple organ dysfunction, and mortality across these studies. Six of these studies focused on or separately analyzed children

Page 30: Surviving Sepsis Campaign International Guidelines for the ... · In 2001, the Surviving Sepsis Campaign (SSC) was formed by the Society of Critical Care Medicine (SCCM), European

Copyright © 2020 by the Society of Critical Care Medicine and the World Federation of Pediatric Intensive and Critical Care Societies.Unauthorized reproduction of this article is prohibited

Online Special Article

Pediatric Critical Care Medicine www.pccmjournal.org e81

with sepsis (433–437). Three studies reported a greater need for vasoactive agents in VDD children (433–435), although mortality across these six studies was not associated with VDD (433–437) (Supplemental Table 26, Supplemental Digital Content 1, http://links.lww.com/PCC/B138).

Vitamin D levels are lowered by fluid resuscitation, which can confound the association with illness severity and disease com-plications (434). In addition, free or bioavailable 1,25(OH)

2D

is the active form which is influenced by the level of vitamin D binding protein (VDBP) and a patient’s VDBP genotype, which was not estimated or measured in prior studies (438). Although vitamin D levels are a potentially modifiable risk factor via sup-plementation, a meta-analysis of rapid normalization of vi-tamin D levels concluded that it is best achieved using loading therapy that takes into account disease status, determines base-line vitamin D level, and considers patient weight (439–441). A loading dose greater than 300,000 international units should be avoided outside of RCTs evaluating risk and benefit.

Hypervitaminosis D is associated with hypercalcemia and other severe complications (442) and vitamin D overdoses can be fatal (443). No current data support that rapid acute correction of VDD is an effective treatment in septic shock or improves outcomes of septic children. Further, measurement of 25(OH)

2D levels is not currently a standard component of

sepsis care and methods of accurately measuring bioavailable vitamin D are not yet widely validated. However, if VDD is diagnosed, repletion should occur as a usual part of general holistic pediatric care according to recommended guidelines independently of the presence of sepsis (444).

BLOOD PRODUCTS

65) We suggest against transfusion of RBCs if the blood he-moglobin concentration is greater than or equal to 7 g/dL in hemodynamically stabilized children with septic shock or other sepsis-associated organ dysfunction (weak recom-mendation, low quality of evidence).

Remarks: According to the 2018 Transfusion and Anemia Expertise Initiative (TAXI) guidelines, for the purposes of RBC transfusion, “hemodynamically stabilized” is defined as a MAP higher than 2 sds below normal for age and no increase in vas-oactive medications for at least 2 hours.

66) We cannot make a recommendation regarding hemo-globin transfusion thresholds for critically ill children with unstable septic shock.

Rationale: The only study evaluating specific RBC trans-fusion thresholds in children with sepsis is a post hoc sub-group analysis of the Transfusion Requirements in the PICU (TRIPICU) study (445) (Supplemental Table 27, Supplemental Digital Content 1, http://links.lww.com/PCC/B138). This study included 137 stabilized critically ill children (MAP > 2 sds below normal for age and cardiovascular support not increased for at least 2 hr before enrollment) with sepsis, with a hemoglobin less than or equal to 9.5 g/dL within 7 days after PICU admis-sion. Patients were randomized to receive RBCs if hemoglobin

decreased to either less than 7.0 g/dL (restrictive group) or 9.5 g/dL (liberal group). No differences were found between the re-strictive versus liberal group in the primary endpoint of new or progressive multiple organ dysfunction syndrome (18.8% vs 19.1%) or mortality (p = 0.44). These results are similar to those from primary analysis of the TRIPICU study (446), as well as in adults (447). Our suggestion against transfusion if hemoglobin is greater than 7 g/dL in hemodynamically stable children with sepsis parallels the TAXI recommendations (448).

Insufficient data are available to guide RBC transfusion therapy in children with unstable septic shock. Two pediatric RCTs did demonstrate decreased mortality when red blood transfusion to goal hemoglobin greater than or equal to 10 (hematocrit > 30%) was included as part of an EGDT algo-rithm targeting Scvo

2, but the impact of each individual com-

ponent, including red blood transfusion, is unclear (190, 449). In critically ill adults, the Transfusion Requirements in Septic Shock trial randomized 998 subjects with septic shock to ei-ther a transfusion threshold hemoglobin of 7 g/dL or 9 g/dL (450). At randomization, all patients had hypotension (mean arterial pressure < 70 mm Hg) and/or were being treated with vasopressors. Ninety-day mortality showed no differences (RR, 0.94; 95% CI, 0.78–1.09), suggesting that a restrictive transfu-sion strategy in hemodynamically unstable septic adults was safe (Supplemental Table 27, Supplemental Digital Content 1, http://links.lww.com/PCC/B138). The SSC recommends that RBC transfusion in adults occur only when hemoglobin concentration decreases to less than 7.0 g/dL in the absence of extenuating circumstances, such as myocardial ischemia, severe hypoxemia, or acute hemorrhage (strong recommen-dation, high quality of evidence) (50). This adult recommen-dation is also valid for hemodynamically unstable patients.

However, in the absence of pediatric data, we are not able to provide a recommendation for critically ill children with un-stable septic shock.

67) We suggest against prophylactic platelet transfusion based solely on platelet levels in nonbleeding children with septic shock or other sepsis-associated organ dysfunction and thrombocytopenia (weak recommendation, very low quality of evidence).

Rationale: One observational study demonstrated an as-sociation between the administration of platelet transfusions to critically ill children and worse clinical outcomes (Sup-plemental Table 28, Supplemental Digital Content 1, http://links.lww.com/PCC/B138), including longer ICU LOS, pro-gressive organ dysfunction, and increased mortality (451). In-direct evidence can be found in an RCT of 660 infants born at less than 34 weeks gestational age, the majority of whom were treated for sepsis, that compared a platelet transfusion threshold of 50,000/mm3 (high threshold) with 25,000/mm3 (low threshold)(452). More infants in the high- versus low-threshold group received at least one platelet transfusion (90% vs 53%). More adverse events, including new major bleeding or death, were also seen in the high threshold group (OR, 1.57; 95% CI, 1.06–2.32).

Page 31: Surviving Sepsis Campaign International Guidelines for the ... · In 2001, the Surviving Sepsis Campaign (SSC) was formed by the Society of Critical Care Medicine (SCCM), European

Copyright © 2020 by the Society of Critical Care Medicine and the World Federation of Pediatric Intensive and Critical Care Societies.Unauthorized reproduction of this article is prohibited

Copyright © 2020 by the Society of Critical Care Medicine and the World Federation of Pediatric Intensive and Critical Care Societies.Unauthorized reproduction of this article is prohibited

Weiss et al

e82 www.pccmjournal.org February 2020 • Volume 21 • Number 2

Although existing evidence does not support a platelet threshold at which transfusion is absolutely indicated, the risk of spontaneous bleeding may be greater at lower platelet counts, for example, less than 10–20,000/mm3. In addition, some populations of thrombocytopenic critically ill children may have a relatively high risk of bleeding, such as those with oncological diagnoses or those receiving ECMO. Because the threshold at which the benefits of platelet transfusion out-weigh the risks is unknown, clinical judgment based on patient risk factors for bleeding in addition to the measured platelet level must be exercised carefully.

68) We suggest against prophylactic plasma transfusion in nonbleeding children with septic shock or other sepsis-associated organ dysfunction and coagulation abnormali-ties (weak recommendation, very low quality of evidence).

Remarks: Prophylactic plasma transfusion refers to situa-tions in which there is an abnormality in laboratory coagula-tion testing but no active bleeding.

Rationale: No direct data exist to inform a recommenda-tion about plasma transfusion in pediatric sepsis. One RCT evaluates prophylactic plasma transfusion in critically ill chil-dren without sepsis. Pieters et al (453) randomized 81 children less than 2 years old requiring primary repair of craniosynosto-sis to receive plasma using either a prophylactic (in absence of bleeding) or reactive (when the patient was bleeding) strategy. The prophylactic plasma transfusion group received a signif-icantly higher volume of plasma compared with the reactive group (29.7 vs 16.1 mL/kg; p < 0.001). Despite an improvement in coagulation values in the prophylactic group, there was no difference in packed RBC transfusion requirements or blood loss between the two groups (Supplemental Table 29, Supple-mental Digital Content 1, http://links.lww.com/PCC/B138). Additionally, a meta-analysis published in 2012 that included 80 RCTs (mostly in adults) concluded that there was no con-sistent evidence for benefit of prophylactic plasma transfusion across a range of indications that were evaluated (454). Obser-vational studies in critically ill children have shown that plasma transfusions are associated with worse clinical outcomes (455, 456). Furthermore, plasma transfusion frequently fails to cor-rect abnormal coagulation tests in critically ill adults and chil-dren (456, 457). We therefore suggest against prophylactic plasma transfusions for children with septic shock and other sepsis-associated organ dysfunction who are not bleeding.

However, some specific patient populations might benefit from prophylactic plasma transfusions, such as patients with worsening coagulation tests at high risk for disseminated in-travascular coagulopathy, children with comorbid cancer, or children with sepsis on extracorporeal life support.

PLASMA EXCHANGE, RENAL REPLACEMENT, AND EXTRACORPOREAL SUPPORT

69) We suggest against using plasma exchange (PLEX) in chil-dren with septic shock or other sepsis-associated organ dys-function without thrombocytopenia-associated multiple

organ failure (TAMOF) (weak recommendation, very low quality of evidence).

70) We cannot suggest for or against the use of PLEX in chil-dren with septic shock or other sepsis-associated organ dysfunction with TAMOF.

Rationale: Therapeutic PLEX for septic shock or sepsis-associated organ dysfunction aims to normalize the plasma milieu of a systemically inflamed septic patient. Currently, no large RCTs have evaluated PLEX in pediatric septic shock or sepsis-associated organ dysfunction. Rimmer et al (458) per-formed a meta-analysis that included four small RCTs evaluat-ing PLEX in adults (n = 128) and pediatric (n = 66) patients with sepsis and septic shock. PLEX was associated with reduced mortality in adults (RR, 0.63; 95% CI, 0.42–0.96), but not in children (RR, 0.96; 95% CI, 0.28–3.38). However, because of the heterogeneity of the patient population, inclusion criteria, technical modalities of PLEX (filtration vs centrifugation), and types of replacement fluid (plasma vs albumin) in these four studies as well as the costs and potential risks, PLEX cannot be routinely recommended as this time (Supplemental Table 30, Supplemental Digital Content 1, http://links.lww.com/PCC/B138). Similarly, the American Society for Apheresis recom-mended that the “optimum role of apheresis therapy is not es-tablished” in sepsis with multiple organ failure (459).

TAMOF is an inflammatory phenotype of sepsis-induced multiple organ dysfunction in children that can be identified clinically by new-onset thrombocytopenia and evolving mul-tiple organ dysfunction (460, 461). Autopsies performed on patients who died with TAMOF revealed disseminated micro-vascular thromboses in various organs (460). These patients had deficient activity of a disintegrin and metalloproteinase with thrombospondin type 1 motif (ADAMTS-13), elevated von Willebrand factor (VWF) activity, and the presence of ultra-large plasma VWF (460, 462). Decreased activity of ADAMTS-13 leads to high circulating levels of ultra-large VWF that induce widespread platelet activation and thrombotic microangiopathy. A number of inflammatory mediators are el-evated in sepsis that can inhibit or inactivate ADAMTS-13 in-cluding interleukin-6, granulocyte elastase, plasmin, thrombin, cell-free hemoglobin, Shiga toxins, and immunoglobulin G auto-antibody (463–468).

Three studies have examined the utility of PLEX in children with sepsis and TAMOF (460, 469, 470). In the most recent and largest study (n = 81), Fortenberry et al (469) reported that PLEX was associated with lower 28-day mortality by mul-tivariate analysis (adjusted RR, 0.45; 95% CI, 0.23–0.90) and by propensity score weighting (adjusted RR, 0.46; 95% CI, 0.22–0.97). In a retrospective cohort study from the Turkish TAMOF Network (n = 42), PLEX was associated with lower 28-day mortality compared with the no PLEX group (27% vs 70%; p = 0.004) [470]). In the third study, Nguyen et al (460) randomized 10 children to either PLEX or standard therapy. The 5 patients who received PLEX had restora-tion of ADAMTS-13 activity and greater survival (5/5) com-pared with standard therapy (1/5; p < 0.05). Taken together, these data support a biologic rationale for the use of PLEX in

Page 32: Surviving Sepsis Campaign International Guidelines for the ... · In 2001, the Surviving Sepsis Campaign (SSC) was formed by the Society of Critical Care Medicine (SCCM), European

Copyright © 2020 by the Society of Critical Care Medicine and the World Federation of Pediatric Intensive and Critical Care Societies.Unauthorized reproduction of this article is prohibited

Online Special Article

Pediatric Critical Care Medicine www.pccmjournal.org e83

TAMOF, that is, the removal of pathologic ultra-large VWF and ADAMTS-13 inhibitors and restoration of ADAMTS-13 activity. This approach of using PLEX is similar to the rationale for using PLEX in thrombotic thrombocytopenic purpura (471). Although the panel acknowledges a potential benefit for PLEX and encourages an RCT to better define the utility of PLEX in children with sepsis and TAMOF, a recommendation could not be made based on existing data.

71) We suggest using renal replacement therapy to prevent or treat fluid overload in children with septic shock or other sepsis-associated organ dysfunction who are unresponsive to fluid restriction and diuretic therapy (weak recommen-dation, very low quality of evidence).

Rationale: Renal replacement therapy is increasingly being used in PICUs for renal and nonrenal conditions. The ra-tionale for renal replacement therapy in septic shock includes impending or established fluid overload following initial re-suscitation or for cytokine removal, reversal of coagulopathy, to buffer lactic acidosis, to address AKI, or a combination of these factors. CRRT may be useful for treating established fluid overload or to prevent further fluid overload while allowing liberal volume administration for nutrition, antimicrobials, and other medications, sedation, and transfusions. In addition, certain techniques of continuous blood purification may help to regulate systemic inflammation and promote kidney re-covery (472). Fluid overload has been shown to cause increased morbidity and mortality in various intensive care settings, and there is documented favorable association of CRRT in fluid overload (473).

However, no high-quality studies in critically ill children with sepsis exist to directly determine whether RRT is de-finitively beneficial compared with diuretics and/or fluid re-striction. Most of the data come from adult studies where outcomes have varied from mortality to ICU LOS and venti-lator- and vasoactive-free days. One study addressed the tim-ing of CRRT initiation in 27 children with sepsis and multiple organ dysfunction, demonstrating that CRRT was associated with survival when started within 48 hours of admission compared with those started on CRRT after 48 hours of ad-mission (61% vs 33%; p < 0.001). However, timing of CRRT initiation was at the discretion of the treating team, raising concern for confounding between groups, and all patients in both groups experienced normalization of kidney function (474) (Supplemental Table 31, Supplemental Digital Content 1, http://links.lww.com/PCC/B138).

The possible benefits of CRRT must also be weighed against potential risks, including the need for an invasive catheter, costs, limited availability in some centers, the need for clinician and nursing-specialist expertise, and the challenge of optimal timing (e.g., following resuscitation for fluid removal or earlier for acute cytokine clearance). Therefore, as the initial treat-ment strategy, we judge that fluid restriction and use of diuret-ics are reasonable in the presence of impending or established fluid overload with CRRT reserved as a second-line option to prevent or treat fluid overload in children with septic shock or

other sepsis-associated organ dysfunction who are unrespon-sive to fluid restriction and diuretic therapy.

72) We suggest against high-volume hemofiltration (HVHF) over standard hemofiltration in children with septic shock or other sepsis-associated organ dysfunction who are treated with renal replacement therapy (weak recommen-dation, low quality of evidence).

Rationale: HVHF for critically ill patients with septic shock and AKI is an appealing strategy for maintaining acid-base and fluid homeostasis, or for having a potential immunomodula-tory effect in sepsis by removal of toxins and other inflamma-tory mediators, especially cytokines that contribute to organ injury and dysfunction.

In adults, use of higher CRRT flux rates (> 35 mL/kg/hr filtration-dialysis), while initially encouraging, has not shown overall mortality benefit in subsequent RCTs and meta-anal-ysis. A 2017 Cochrane review found no significant benefit in mortality, severity of organ dysfunction, LOS, or adverse effects with HVHF versus standard hemofiltration rates in critically ill adults (475). Notably, the results of this meta-analysis show that very few studies have been conducted to investigate the use of HVHF in critically ill patients with septic shock (four stud-ies totaling 201 participants).

In a study involving 155 pediatric patients with severe sepsis, HVHF treatment did not significantly reduce 28-day mortality compared with standard volume CRRT. Furthermore, there were no significant reductions in plasma levels of inflamma-tory mediators or in improving hemodynamic variables for HVHF. However, the incidence of hyperglycemia was signif-icantly higher in HVHF group than in CVVH group (476) (Supplemental Table 32, Supplemental Digital Content 1, http://links.lww.com/PCC/B138).

73) We suggest using venovenous ECMO in children with sep-sis-induced PARDS and refractory hypoxia (weak recom-mendation, very low quality of evidence).

Rationale: ECMO was introduced more than 40 years ago to support patients with reversible but severe cardiovascular and/or respiratory failure refractory to conventional medical therapy. As such, children with life-threatening sepsis-induced ARDS are often considered as candidates for ECMO rescue (477), and PALICC endorsed ECMO for the treatment of re-fractory hypoxia (24). The use of ECMO in pediatric sepsis has increased over the past decade (478, 479); whether this has improved survival remains to be determined (480). To date, no RCT examining the effect of ECMO on outcome in pedi-atric sepsis has been published. In the absence of such data, using propensity score matching, Barbaro et al (481) reported that children with severe PARDS enrolled in the RESTORE trial had similar mortality rates when supported with ECMO (15/61, 25%) as compared with those who were not (18/61, 30%) (482) (Supplemental Table 33, Supplemental Digital Content 1, http://links.lww.com/PCC/B138). Research is un-derway to determine optimal pre-ECMO candidacy (483) as measures of renal, hepatic, neurologic, and hematologic

Page 33: Surviving Sepsis Campaign International Guidelines for the ... · In 2001, the Surviving Sepsis Campaign (SSC) was formed by the Society of Critical Care Medicine (SCCM), European

Copyright © 2020 by the Society of Critical Care Medicine and the World Federation of Pediatric Intensive and Critical Care Societies.Unauthorized reproduction of this article is prohibited

Copyright © 2020 by the Society of Critical Care Medicine and the World Federation of Pediatric Intensive and Critical Care Societies.Unauthorized reproduction of this article is prohibited

Weiss et al

e84 www.pccmjournal.org February 2020 • Volume 21 • Number 2

dysfunction, and particularly the presence of bloodstream infections, seem to discriminate mortality risk better than traditional pediatric severity of illness scores such as PRISM, Pediatric Index of Mortality, and Pediatric Logistic Organ Dysfunction. Clearly, ECMO is not available worldwide, and transfer of highly unstable patients to higher levels of care that offer the therapy can carry substantial risk. However, adult and pediatric data suggest a potential association with improved mortality, particularly if transfer is to high volume ECMO centers (484, 485).

74) We suggest using venoarterial ECMO as a rescue therapy in children with septic shock only if refractory to all other treatments (weak recommendation, very low quality of evidence).

Rationale: Several anecdotal reports of use of venoarte-rial ECMO in the management of RSS in children exist. More recent reports suggest that venoarterial ECMO may be as-sociated with better survival than conventional therapy, and strategies to maximize flow rates to reverse shock and mul-tiple organ dysfunction may play an important role (486, 487). However, considerable concern surrounds the risks of this highly invasive therapy, such as hemorrhage and throm-boembolic events.

The most recent and largest report of venoarterial ECMO in 44 pediatric patients with RSS secondary to bacterial, viral, or fungal infection admitted to seven tertiary PICUs across five different countries compared their outcome to 120 children with RSS managed by conventional therapy (488). Inclusion in the study required children to meet three of four criteria for severe septic shock in the first 24 hours of their ICU stay: arterial pH less than or equal to 7.15, arterial lactate greater than or equal to 4.0 mmol/L, base excess less than or equal to –10 mmol/L, and in-hospital cardiac arrest. Patients were excluded if they had cyanotic congenital heart disease, myocar-ditis, or an out-of-hospital cardiac arrest. The results showed no significant difference in survival to hospital discharge (50% in the venoarterial ECMO cohort vs 40% in the conventional therapy cohort). Survival was significantly higher in patients who received high ECMO flows (> 150 mL/kg/min at 4 hr after institution of ECMO) compared with children who received standard ECMO flows or no ECMO (Supplemental Table 34, Supplemental Digital Content 1, http://links.lww.com/PCC/B138).

The potential use of venoarterial ECMO for RSS suggests that the definition of RSS should be standardized across insti-tutions. As yet, no universal definition of RSS in children exists. One published definition that could be applied is from the European Society of Paediatric and Neonatal Intensive Care (10). The suggested definition for RSS was blood lac-tate greater than 8 mmol/L or a 1 mmol/L lactate increase after 6 hours of resuscitation and high vasoactive dependency (vasopressor-inotrope score > 200), or myocardial dysfunc-tion defined as the occurrence of a resuscitation-responsive cardiac arrest in PICU or cardiac ultrasound findings with left

ventricle ejection fraction less than 25% or a cardiac index less than 2.2 L/min/m2.

IMMUNOGLOBULINS

75) We suggest against the routine use of IV immune glob-ulin (IVIG) in children with septic shock or other sepsis-associated organ dysfunction (weak recommendation, low quality of evidence).

Remarks: Although routine use of IVIG is not recom-mended, select patients may benefit from such treatment.

Rationale: The proposed rationale for IVIG in severe infections is to boost passive immunity through neutraliza-tion of bacterial toxins, promoting opsonization of bacteria, and inhibition of immune cell proliferation and inflamma-tory mediators. However, IVIG has considerable batch-to-batch variability and its true biologic activity is not clear. There are no high-quality studies of IVIG in critically ill children with sepsis, and small observational studies have re-ported conflicting results (489). An RCT of polyclonal IVIG in 100 children with sepsis demonstrated a reduction in mor-tality (28% vs 44%), LOS (6 vs 9 d), and less progression to complications (8% vs 32%) (490). However, a more recent multicenter trial of polyclonal IVIG in 3,493 neonates with suspected or proven serious infection found no significant differences in mortality or major disability (491). Other stud-ies have been carried out with specific monoclonal antibod-ies (e.g., monoclonal antibody against endotoxin in children with meningococcal septic shock), but there are no definitive data to support general benefit of polyclonal immunoglob-ulin in neonates or children with septic shock at this time. Data from adult patients with septic shock also do not sup-port a routine benefit of IVIG (492), although administration of immunoglobulin M- and immunoglobulin A-enriched polyclonal IVIG has shown possible efficacy (493) (Supple-mental Table 35, Supplemental Digital Content 1, http://links.lww.com/PCC/B138).

For patients with toxic shock syndrome, especially those with streptococcal etiology, polyclonal IVIG may have clinical utility (494). Other potential pediatric populations that may benefit from IVIG in sepsis are those with necrotizing fasciitis (although evidence in adults does not support use [495, 496]), and those with primary humoral immunodeficiencies or im-munocompromised with documented low immunoglobulin levels.

PROPHYLAXIS

76) We suggest against the routine use of stress ulcer prophy-laxis in critically ill children with septic shock or other sepsis-associated organ dysfunction, except for high-risk patients (weak recommendation, very low quality of evidence).

Remarks: Although routine stress-ulcer prophylaxis is not recommended, some high-risk patients may benefit from

Page 34: Surviving Sepsis Campaign International Guidelines for the ... · In 2001, the Surviving Sepsis Campaign (SSC) was formed by the Society of Critical Care Medicine (SCCM), European

Copyright © 2020 by the Society of Critical Care Medicine and the World Federation of Pediatric Intensive and Critical Care Societies.Unauthorized reproduction of this article is prohibited

Online Special Article

Pediatric Critical Care Medicine www.pccmjournal.org e85

stress ulcer prophylaxis. Studies have supported benefit of stress-ulcer prophylaxis when baseline rate of clinically impor-tant bleeding is approximately 13%.

Rationale: Stress ulcer prophylaxis should not be rou-tinely administered to children with septic shock or other sepsis-associated organ dysfunction, as evidence for benefit is lacking (497) and may increase risk of adverse effects, such as pneumonia or C. difficile (formerly Clostridium) infec-tion (498). Rather than routine, universal administration of stress-ulcer prophylaxis, individual patients should be assessed for the presence of risk factors of clinically impor-tant gastrointestinal bleeding. These include multiple organ dysfunction (499), prolonged mechanical ventilation (> 48 hr), coagulopathy, persistent shock, and treatment with corticosteroids and nonsteroidal anti-inflammatory agents (500).

The risk of GI bleeding is also reduced by mucosal pro-tection introduced by gastric feeding. Early enteral nutrition could therefore be a viable alternative to pharmacological stress-ulcer prophylaxis. A meta-analysis of 1,836 adult patients reported that, in the presence of enteral nutri-tion, pharmacological stress ulcer prophylaxis did not sig-nificantly change the risk of GI bleeding. Notably, in those patients who received enteral nutrition and were treated with stress ulcer prophylaxis, the risk of pneumonia was increased compared with patients not treated with stress ulcer prophy-laxis (OR, 2.81; 95% CI, 1.2–6.6) (501) (Supplemental Table 36, Supplemental Digital Content 1, http://links.lww.com/PCC/B138).

77) We suggest against routine deep vein thrombosis (DVT) prophylaxis (mechanical or pharmacologic) in critically ill children with septic shock or other sepsis-associated organ dysfunction, but potential benefits may outweigh risks and costs in specific populations (weak recommendation, low quality of evidence).

Rationale: An open-label RCT of low molecular weight hep-arin to prevent CVC-associated thrombosis in the PICU was terminated early because of poor recruitment (502). Eleven of 78 patients (14.1%) randomized to reviparin had DVT proven on venogram versus 10 of 80 controls (12.5%) (OR, 1.15; 95% CI, 0.42–3.23). Three adverse events (major bleed or death) all occurred in the control group and no deaths occurred because of venous thromboembolism (Supplemental Table 37, Sup-plemental Digital Content 1, http://links.lww.com/PCC/B138). A subsequent systematic review found the quality of evidence to be low and that the efficacy of low molecular weight heparin in preventing CVC-associated thrombosis is unknown (503). It is important to highlight that these studies were specific to children with CVCs who may or may not have had sepsis and that they may not apply to the general thromboembolic risk in children with sepsis.

Although CVCs represent the principal risk factor for DVT in infants (504), older children may have other risk factors. For example, the risk of DVT increases in adoles-cence, obesity, cancer, and in those with multiple medical

conditions, especially renal and cardiac disease (505, 506). At present, it is unknown whether certain high-risk pop-ulations of children with sepsis may benefit from DVT prophylaxis.

KNOWLEDGE GAPS AND RESEARCH OPPORTUNITIESThis report from the SSC pediatric guidelines panel covers five main topic areas (i.e., early recognition and infection, hemodynamics, ventilation, endocrine and metabolic thera-pies, and adjunctive therapies) with a total of 77 recommen-dations arising from 67 PICO questions. On review of these evidence-based analyses, it is clear that, for many PICO ques-tions, the literature review failed to identify sufficient data to develop strong (or even weak in some instances) recommen-dations for critically ill children with septic shock or other sepsis-associated organ dysfunction. These SSC pediatric guidelines, at the same time, also identified gaps that can in-form future research opportunities. As new research populates the evidence-base, it can then be used to develop future itera-tions of the SSC pediatric guidelines, creating a cycle designed to grow the evidence and increase the number of strong rec-ommendations in the future. Further clarity is needed from both informative pathophysiology studies as well as well-designed RCTs, and the panelists have listed these in the text. The design of meaningful and effective future research should be informed by the needs identified by the collective clinical expertise within the panel.

Overall, the process of developing the SSC-pediatric guide-lines generated at least 29 pathophysiology questions warrant-ing further study and 23 RCTs (i.e., total of 52 studies). We present these questions as research opportunities, but have not yet prioritized these opportunities into a formal research agenda (Table 6). We envision that many of the pathophysi-ology questions can be taken up by individual research groups, and we hope that the SSC children’s guidelines document will serve as a template of current evidence and how best to fill the gaps in our knowledge. In contrast, the necessary RCTs will need a coordinated national/international effort and our com-munity will need to prioritize the most appropriate studies at different phases of management (i.e., recognition, fluid resus-citation, first 48 hr).

ACKNOWLEDGMENTSWe wish to thank Darlene Barkman, MA, and Janna Pogers, PT, MPT, NCS, CSRS for their sensitive and insightful com-ments from the perspective of parents of children with sepsis. Their input, particularly related to ranking the importance of outcomes to consider through the literature search, pro-vided valuable direction to the panel. We also wish to thank Rebecca Skidmore and James D. Medd for their dedication as they conducted the literature searches for the five panels. Their experience and professionalism contributed greatly to the final publication. Finally, appreciation is extended to Deborah L. McBride for project management and editorial support.

Page 35: Surviving Sepsis Campaign International Guidelines for the ... · In 2001, the Surviving Sepsis Campaign (SSC) was formed by the Society of Critical Care Medicine (SCCM), European

Copyright © 2020 by the Society of Critical Care Medicine and the World Federation of Pediatric Intensive and Critical Care Societies.Unauthorized reproduction of this article is prohibited

Copyright © 2020 by the Society of Critical Care Medicine and the World Federation of Pediatric Intensive and Critical Care Societies.Unauthorized reproduction of this article is prohibited

Weiss et al

e86 www.pccmjournal.org February 2020 • Volume 21 • Number 2

TABLE 6. Knowledge Gaps and Research Opportunities (Refer to Numbered Recommendations in Guidelines and Appendix 1)

Subgroup Pathophysiology Clinical Trials

Screening, diagnosis, and systematic management of sepsis: Four pathophysiology studies and two RCTs

QI screening tool algorithms to recognize clinical deterioration (see Rec 1)

Define the optimal level of hyperlactatemia (see Rec 2)

Protocol/guideline for management (see Rec 3)New molecular technologies in identifying pathogens

before blood culture positivity or after antibiotic administration (see Rec 4)

Pediatric sepsis recognition (see Rec 1)

Initial or serial measurement of blood lactate directly informs evaluation and/or management (see Rec 2)

Antimicrobial therapy: Seven pathophysiology studies

The definition of “timely” antimicrobials in a bundle of initial care (see Recs 5 and 6)

QI metrics to assess unnecessary antimicrobials (see Rec 7)

Antimicrobial resistance rate thresholds to help decide when the addition of a glycopeptide or second gram-negative agent is necessary (see Rec 10 and 11)

Alteration in the pharmacokinetics and pharmacodynamics of antimicrobials (see Rec 12)

The relationship between antimicrobial stewardship programs and a decrease in antimicrobial resistance (see Rec 13)

The use of procalcitonin as a guide to antimicrobial therapy and relationship to outcome (see Rec 13)

The determinants of optimal duration of antimicrobial therapy (see Rec 14)

Source control: One pathophysiology studies

Role of source control (see Rec 15 and 16)

Fluid therapy: One pathophysiology study and two RCTs

Features of early recognition of fluid overload (see Rec 17–19)

Clinical markers of cardiac output to guide fluid resuscitation (see Rec 17–19)

Balanced crystalloid vs 0.9% saline (see Rec 21)

Hemodynamic monitoring: Two RCTs

Specific hemodynamic targets (> 5th vs > 50th mean arterial pressure percentile) (see Rec 24)

Lactate-guided resuscitation (see Rec 27)

Vasoactive medications: Three pathophysiology studies

The choice of first-line vasoactive infusion (see Rec 30)

The optimal threshold for using continuous infusion of vasopressin (see Rec 32)

The use and effects of inodilators (see Rec 33)

(Continued )

Page 36: Surviving Sepsis Campaign International Guidelines for the ... · In 2001, the Surviving Sepsis Campaign (SSC) was formed by the Society of Critical Care Medicine (SCCM), European

Copyright © 2020 by the Society of Critical Care Medicine and the World Federation of Pediatric Intensive and Critical Care Societies.Unauthorized reproduction of this article is prohibited

Online Special Article

Pediatric Critical Care Medicine www.pccmjournal.org e87

TABLE 6. (Continued). Knowledge Gaps and Research Opportunities (Refer to Numbered Recommendations in Guidelines and Appendix 1)

Subgroup Pathophysiology Clinical Trials

Ventilation: Four pathophysiology studies and five RCTs

Noninvasive modalities to identify the need for early mechanical ventilation (see Rec 36)

Does early noninvasive mechanical ventilation vs invasive mechanical ventilation in sepsis-induced PARDS mitigate the need for subsequent invasive mechanical ventilation (see Rec 36)

The optimal approach to setting PEEP in mechanical ventilation for sepsis-induced PARDS (see Rec 37)

The use of recruitment maneuvers in mechanical ventilation for sepsis-induced PARDS (see Rec 38)

Early- vs delayed-endotracheal intubation for refractory shock without respiratory failure (see Rec 34)

During mechanical ventilation, low- vs moderate-PEEP strategy for sepsis-induced PARDS (see Rec 37)

During mechanical ventilation, prone positioning in sepsis-induced severe PARDS (current RCT will need secondary analysis of this subgroup) (see Rec 39)

High-frequency oscillatory ventilation vs conventional mechanical ventilation in sepsis-induced severe PARDS (current RCT will need secondary analysis of this subgroup) (see Rec 42)

Neuromuscular blocking agent during mechanical ventilation for sepsis-induced severe PARDS (current RCT will need secondary analysis of this subgroup) (see Rec 43)

Corticosteroids: One RCT Adjunctive corticosteroids for refractory septic shock (see Rec 45)

Endocrine and metabolic therapies: Two pathophysiology studies and one RCT

The optimal glucose target (between 140 and 180 mg/dL) necessitating control with insulin in children with septic shock or other sepsis-associated organ dysfunction (see Rec 47)

Hypocalcemia and supplements in children with septic shock or other sepsis-associated organ dysfunction (see Rec 48)

Fever management in children with septic shock or other sepsis-associated organ dysfunction (see Rec 50)

Nutrition: Three pathophysiology studies and seven RCTs

Lipid solution effects on inflammatory physiology (see Rec 54)

The role of prokinetic agents in immunocompromised children with septic shock or other sepsis-associated organ dysfunction (see Rec 57)

The prevalence of low serum vitamin C levels in children with septic shock or other sepsis-associated organ dysfunction (see Rec 62)

Early- vs late- enteral nutrition in children with septic shock or other sepsis-associated organ dysfunction (see Recs 51, 52, and 53)

Bolus vs continuous enteral feeding in children with septic shock or other sepsis-associated organ dysfunction (see Rec 51, 52, and 53)

Enteral nutrition vs parenteral supplementation of nutritional intake in the first 7 d of management of children with septic shock or other sepsis-associated organ dysfunction (see Rec 53)

Dietary supplements (selenium, glutamine, arginine, zinc) in children with septic shock or other sepsis-associated organ dysfunction (see Recs 58, 59, 60, and 61)

Vitamin C supplementation in children with septic shock or other sepsis-associated organ dysfunction (see Rec 62)

Thiamine deficiency and supplementation in children with septic shock or other sepsis-associated organ dysfunction (see Rec 63)

Vitamin D deficiency and supplementation in children with septic shock or other sepsis-associated organ dysfunction (see Rec 64)

(Continued )

Page 37: Surviving Sepsis Campaign International Guidelines for the ... · In 2001, the Surviving Sepsis Campaign (SSC) was formed by the Society of Critical Care Medicine (SCCM), European

Copyright © 2020 by the Society of Critical Care Medicine and the World Federation of Pediatric Intensive and Critical Care Societies.Unauthorized reproduction of this article is prohibited

Copyright © 2020 by the Society of Critical Care Medicine and the World Federation of Pediatric Intensive and Critical Care Societies.Unauthorized reproduction of this article is prohibited

Weiss et al

e88 www.pccmjournal.org February 2020 • Volume 21 • Number 2

Drs. Weiss and Peters served as arbiters for conflict interest management and adjudication throughout the guidelines process following standard operating procedures set forth by Society of Critical Care Medicine (SCCM) and endorsed by European Society of Intensive Care Medicine. Dr. Weiss participates in Pediatric Acute Lung Injury and Sepsis Inves-tigators (PALISI) and Shock Society. Dr. Peters participates in the UK PICS study group (vice-chair) and has testified as an expert witness in cases of clinical negligence, causation of injuries. Dr. Agus participates in the American Academy of Pediatrics (AAP), Pediatric Academic Societies (PAS), American Pediatric Society, Society for Pediatric Research, and The American Society for Clinical Investigation, and he has testified as an expert witness in cases related to ICU and/or endocrinology in children. Dr. Flori participates in American Thoracic Society (ATS) State Chapter (Executive Board Member—Michigan and California State Chapters) and PALISI Network, Grant funding from Gerber Foundation (Steering commit-tee member for various studies being implemented through the Network from intramural funding, governmental or other Foundation Grant fund-ing). Dr. Nadel received funding form La Jolla Pharmaceutical (consulting), and he participates in the European Society of Pediatric and Neonatal Intensive Care (ESPNIC) (Medical President). Dr. Brierley participates in the ESPNIC. Dr. Carrol participates in National Institute for Health and Care Excellence (NICE) (Diagnostic Advisory Committee panel) and Na-tional Institutes for Health Research (two scientific panels, i4i and DTF). Dr. Cheifetz participates in American Association for Respiratory Care and ATS (volunteer activities) and has testified as an expert witness for medical malpractice cases, he is an advisor to Philips, and a contributor to Up-to-Date. Dr. Cies received funding from Allergan, Merck, Thermo Fisher Scientific, and Atlantic Diagnostic Laboratories (consultant), and he participates in Pediatric Pharmacy Advocacy Group (multiple posi-tions), Society of Infectious Diseases Pharmacists (Vice-Chair of the Inter-organizations Liaison Committee), and the American College of Clinical Pharmacists (member and fellow). Dr. Cruz has testified as an expert wit-ness in cases of children with tuberculosis-related meningitis and is an associate editor for Pediatrics. Dr. De Luca serves as Medical President-e-lect on the Executive Committee of ESPNIC, he served as a consultant and lecturer on the external advisory board and received research and educational grants from Chiesi Farmaceutici S.p.A and AbbVie, and travel grants from AbbVie, he has been a lecturer for Philips, Radiometer, and Waire. Dr. Faust served as chair of the UK NICE Guideline Committee for Sepsis in Children and Adults published in 2016 and for Lyme disease

published in 2018, serves as a regional representative to the UK NHS England Clinical Reference Group for commissioning pediatric specialist medicine care (immunology and infection). Dr. Hall receives funding from La Jolla Pharmaceuticals (consultant on the data safety monitoring board for a clinical trial of a sepsis therapeutic), and he participates in the ATS (online journal club editor) and the American Board of Pediatrics (Critical Care Medicine sub-board). Dr. Ishimine participates in SAEM (Consensus Conference Co-Chair), American Board of Pediatrics/American Board of Emergency Medicine (Immediate Past Chair of the Pediatric Emergency Medicine Subboard), and the American College of Emergency Physicians (Pediatric Emergency Medicine Committee member). Dr. Javouhey re-ceived funding from CSL Behring (trial on IV Immunoglobulins in toxic shock syndrome in children). Dr. Karam participates in BloodNet, PALISI, ISBT, AABB, and CCCTG. Dr. Kneyber participates in the European So-ciety for Pediatric and Neonatal Intensive Care. Dr. MacLaren participates in Extracorporeal Life Support Organization (Executive Committee). Dr. Mehta participates in the American Society for Parenteral and Enteral Nutrition (president). Dr. Møller participates in the Science Systems and Applications, Inc (board member). Dr. Newth received funding from Philips Research North America (consulting concerning monitoring in PICU), and he participates in the ATS. Dr. Nishisaki’s institutional department receives an unrestricted grant from Nihon Kohden (involves an activity to develop a device to measure capillary refill time), and he participates in the So-ciety for Simulation in Healthcare and International Society for Pediatric Simulation. Dr. Nunnally participates in American College of Critical Care Medicine (Regent), Society of Critical Care Anesthesiologists (director), American Society of Anesthesiologists (committee), International Anes-thesia Research Society, and NYSA. Dr. Randolph’s institution received funding from Genentech (influenza biomarker study research support); she has received funding from Bristol Myers Squibb (consultant in 2017) and La Jolla Pharmaceuticals (design of pediatric septic shock trial of an-giotensin II); and she participates in the ATS and the International Sepsis Forum. Dr. Ranjit participates as the Chancellor of College of Pediatric Critical Care, India. Dr. Tume participates in ESPNIC (Nursing President) and the UK PICS Scientific and Education Committee. Dr. Verger par-ticipates in the American Association of Critical-Care Nurses (Cert. Corp. Governance Committee) and the Academy of Nursing (Acute and Critical Care Special Interest Group). Dr. Williams participates in the Pediatric Cardiac Intensive Care Society. Dr. Wolf received funding support for participation in industry-sponsored research from Merck & Co, Astellas,

Blood products: Two pathophysiology studies

Optimal hemoglobin level in children with septic shock or other sepsis-associated organ dysfunction (see Recs 65 and 66)

The threshold at which the benefits of platelet transfusion outweigh the risks in children with septic shock or other sepsis-associated organ dysfunction (see Rec 67)

Plasma exchange, renal replacement, and extracorporeal support: Two pathophysiology studies and two RCTs

Optimal timing and approach for ECMO in refractory shock (see Rec 74)

To define optimal pre-ECMO candidacy (see Recs 73 and 74)

Plasma exchange in children with septic shock or sepsis-associated organ dysfunction with thrombocytopenia-associated organ failure (see Rec 70)

Renal replacement therapy vs diuretics in the first 48 hr in children with septic shock or other sepsis-associated organ dysfunction (see Recs 71–72)

Immunoglobulins

Prophylaxis: One RCT Stress ulcer prophylaxis in relation to feeding in children with septic shock or other sepsis-associated organ dysfunction (see Rec 76)

ECMO = extracorporeal membrane oxygenation, PARDS = pediatric acute respiratory distress syndrome, PEEP = positive end-expiratory pressure, QI = quality improvement, RCT = randomized clinical trial, Rec = recommendation number.

TABLE 6. (Continued). Knowledge Gaps and Research Opportunities (Refer to Numbered Recommendations in Guidelines and Appendix 1)

Subgroup Pathophysiology Clinical Trials

Page 38: Surviving Sepsis Campaign International Guidelines for the ... · In 2001, the Surviving Sepsis Campaign (SSC) was formed by the Society of Critical Care Medicine (SCCM), European

Copyright © 2020 by the Society of Critical Care Medicine and the World Federation of Pediatric Intensive and Critical Care Societies.Unauthorized reproduction of this article is prohibited

Online Special Article

Pediatric Critical Care Medicine www.pccmjournal.org e89

and Cempra Pharmaceuticals, and he received other support from Karius, Empatica, and Bluespark Technologies. Dr. Zimmerman (Past President of SCCM) received funding from Immunexpress, Seattle (sepsis biomarker research), and he participates in the AAP and PAS. Dr. Tissieres received funding from Baxter acute therapies, Bristol-Myers Squibb Company, Chiesi Farmaceutici S.p.A., Faron Pharmaceuticals (consulting, renal re-placement therapy), and Biomerieux, funding from La Jolla Pharmaceu-ticals, Chiesi Farmaceutici S.p.A., and is President ESPNIC (research grant, biomarkers sepsis), and he participates in the Swiss Intensive Care Society, Swiss Pediatric Society, and the French Society of Intensive Care. The remaining authors have disclosed that they do not have any potential conflicts of interest.

REFERENCES 1. Fleischmann-Struzek C, Goldfarb DM, Schlattmann P, et al: The

global burden of paediatric and neonatal sepsis: A systematic review. Lancet Respir Med 2018; 6:223–230

2. Balamuth F, Weiss SL, Neuman MI, et al: Pediatric severe sepsis in U.S. children’s hospitals. Pediatr Crit Care Med 2014; 15:798–805

3. Odetola FO, Gebremariam A, Freed GL: Patient and hospital corre-lates of clinical outcomes and resource utilization in severe pediatric sepsis. Pediatrics 2007; 119:487–494

4. Ruth A, McCracken CE, Fortenberry JD, et al: Pediatric severe sepsis: Current trends and outcomes from the Pediatric Health Information Systems database. Pediatr Crit Care Med 2014; 15:828–838

5. Weiss SL, Fitzgerald JC, Pappachan J, et al; Sepsis Prevalence, Outcomes, and Therapies (SPROUT) Study Investigators and Pediatric Acute Lung Injury and Sepsis Investigators (PALISI) Network: Global epidemiology of pediatric severe sepsis: The sepsis prevalence, outcomes, and therapies study. Am J Respir Crit Care Med 2015; 191:1147–1157

6. Schlapbach LJ, Straney L, Alexander J, et al; ANZICS Paediatric Study Group: Mortality related to invasive infections, sepsis, and septic shock in critically ill children in Australia and New Zealand, 2002-13: A multicentre retrospective cohort study. Lancet Infect Dis 2015; 15:46–54

7. Ames SG, Davis BS, Angus DC, et al: Hospital variation in risk-adjusted pediatric sepsis mortality. Pediatr Crit Care Med 2018; 19:390–396

8. Evans IVR, Phillips GS, Alpern ER, et al: Association between the New York sepsis care mandate and in-hospital mortality for pediatric sepsis. JAMA 2018; 320:358–367

9. Prout AJ, Talisa VB, Carcillo JA, et al: Children with chronic di-sease bear the highest burden of pediatric sepsis. J Pediatr 2018; 199:194–199.e1

10. Morin L, Ray S, Wilson C, et al; ESPNIC Refractory Septic Shock Definition Taskforce the Infection Systemic Inflammation Sepsis sec-tion of ESPNIC: Refractory septic shock in children: A European Society of Paediatric and Neonatal Intensive Care definition. Intensive Care Med 2016; 42:1948–1957

11. Schlapbach LJ, MacLaren G, Festa M, et al; Australian & New Zealand Intensive Care Society (ANZICS) Centre for Outcomes & Resource Evaluation (CORE) and Australian & New Zealand Intensive Care Society (ANZICS) Paediatric Study Group: Prediction of pediatric sepsis mortality within 1 h of intensive care admission. Intensive Care Med 2017; 43:1085–1096

12. Weiss SL, Balamuth F, Hensley J, et al: The epidemiology of hospital death following pediatric severe sepsis: When, why, and how children with sepsis die. Pediatr Crit Care Med 2017; 18:823–830

13. Cvetkovic M, Lutman D, Ramnarayan P, et al: Timing of death in chil-dren referred for intensive care with severe sepsis: Implications for interventional studies. Pediatr Crit Care Med 2015; 16:410–417

14. World Health Organization: Guideline: Updates on Paediatric Emergency Triage, Assessment and Treatment: Care of Critically-Ill Children. Geneva, Switzerland, World Health Organization, 2016. Available at: https://www.ncbi.nlm.nih.gov/books/NBK350523/. Accessed August 18, 2019

15. Davis AL, Carcillo JA, Aneja RK, et al: The American college of critical care medicine clinical practice parameters for hemodynamic support

of pediatric and neonatal septic shock: Executive summary. Pediatr Crit Care Med 2017; 18:884–890

16. NICE. National Institute for Health and Care Excellence: Sepsis: Recognition, Diagnosis and Early Management (NICE Guideline 51). 2016. Available at: https://www.nice.org.uk/guidance/ng51. Accessed August 18, 2019

17. Goldstein B, Giroir B, Randolph A; International Consensus Conference on Pediatric Sepsis: International pediatric sepsis con-sensus conference: Definitions for sepsis and organ dysfunction in pediatrics. Pediatr Crit Care Med 2005; 6:2–8

18. Singer M, Deutschman CS, Seymour CW, et al: The third international consensus definitions for sepsis and septic shock (Sepsis-3). JAMA 2016; 315:801–810

19. Matics TJ, Sanchez-Pinto LN: Adaptation and validation of a pediatric se-quential organ failure assessment score and evaluation of the sepsis-3 definitions in critically ill children. JAMA Pediatr 2017; 171:e172352

20. Schlapbach LJ, Straney L, Bellomo R, et al: Prognostic accuracy of age-adapted SOFA, SIRS, PELOD-2, and qSOFA for in-hospital mortality among children with suspected infection admitted to the in-tensive care unit. Intensive Care Med 2018; 44:179–188

21. Schlapbach LJ, Kissoon N: Defining pediatric sepsis. JAMA Pediatr 2018; 172:312–314

22. Leteurtre S, Duhamel A, Salleron J, et al; Groupe Francophone de Réanimation et d’Urgences Pédiatriques (GFRUP): PELOD-2: An update of the PEdiatric logistic organ dysfunction score. Crit Care Med 2013; 41:1761–1773

23. Proulx F, Gauthier M, Nadeau D, et al: Timing and predictors of death in pediatric patients with multiple organ system failure. Crit Care Med 1994; 22:1025–1031

24. Pediatric Acute Lung Injury Consensus Conference Group: Pediatric acute respiratory distress syndrome: Consensus recommendations from the pediatric acute lung injury consensus conference. Pediatr Crit Care Med 2015; 16:428–439

25. Higgins JP, Altman DG, Gøtzsche PC, et al; Cochrane Bias Methods Group; Cochrane Statistical Methods Group: The cochrane collabo-ration’s tool for assessing risk of bias in randomised trials. BMJ 2011; 343:d5928

26. Wells G, Shea B, O’Connell D, et al: The Newcastle-Ottawa Scale (NOS) for Assessing the Quality of Nonrandomised Studies in Meta-Analyses. 2009. Available at: http://www.ohri.ca/programs/clinical_epidemiology/oxford.htm. Accessed August 18, 2019

27. Guyatt GH, Oxman AD, Vist GE, et al; GRADE Working Group: GRADE: An emerging consensus on rating quality of evidence and strength of recommendations. BMJ 2008; 336:924–926

28. Neumann I, Brignardello-Petersen R, Wiercioch W, et al: The GRADE evidence-to-decision framework: A report of its testing and applica-tion in 15 international guideline panels. Implement Sci 2016; 11:93

29. Alexander PE, Gionfriddo MR, Li SA, et al: A number of factors ex-plain why WHO guideline developers make strong recommenda-tions inconsistent with GRADE guidance. J Clin Epidemiol 2016; 70:111–122

30. Guyatt GH, Schünemann HJ, Djulbegovic B, et al: Guideline pan-els should not GRADE good practice statements. J Clin Epidemiol 2015; 68:597–600

31. Paul R, Melendez E, Stack A, et al: Improving adherence to PALS septic shock guidelines. Pediatrics 2014; 133:e1358–e1366

32. Paul R, Neuman MI, Monuteaux MC, et al: Adherence to PALS sepsis guidelines and hospital length of stay. Pediatrics 2012; 130:e273–e280

33. Lane RD, Funai T, Reeder R, et al: High reliability pediatric septic shock quality improvement initiative and decreasing mortality. Pediatrics 2016; 138:e20154153

34. Balamuth F, Weiss SL, Fitzgerald JC, et al: Protocolized treatment is associated with decreased organ dysfunction in pediatric severe sepsis. Pediatr Crit Care Med 2016; 17:817–822

35. Akcan Arikan A, Williams EA, Graf JM, et al: Resuscitation bundle in pediatric shock decreases acute kidney injury and improves out-comes. J Pediatr 2015; 167:1301–1305.e1

Page 39: Surviving Sepsis Campaign International Guidelines for the ... · In 2001, the Surviving Sepsis Campaign (SSC) was formed by the Society of Critical Care Medicine (SCCM), European

Copyright © 2020 by the Society of Critical Care Medicine and the World Federation of Pediatric Intensive and Critical Care Societies.Unauthorized reproduction of this article is prohibited

Copyright © 2020 by the Society of Critical Care Medicine and the World Federation of Pediatric Intensive and Critical Care Societies.Unauthorized reproduction of this article is prohibited

Weiss et al

e90 www.pccmjournal.org February 2020 • Volume 21 • Number 2

36. Cruz AT, Perry AM, Williams EA, et al: Implementation of goal-directed therapy for children with suspected sepsis in the emergency depart-ment. Pediatrics 2011; 127:e758–e766

37. Scott HF, Deakyne SJ, Woods JM, et al: The prevalence and diagnostic utility of systemic inflammatory response syndrome vital signs in a pedi-atric emergency department. Acad Emerg Med 2015; 22:381–389

38. Parshuram CS, Dryden-Palmer K, Farrell C, et al; Canadian Critical Care Trials Group and the EPOCH Investigators: Effect of a pedi-atric early warning system on all-cause mortality in hospitalized pe-diatric patients: The EPOCH randomized clinical trial. JAMA 2018; 319:1002–1012

39. Moore LJ, Jones SL, Kreiner LA, et al: Validation of a screening tool for the early identification of sepsis. J Trauma 2009; 66:1539–1546; discussion 1546–1547

40. Sepanski RJ, Godambe SA, Mangum CD, et al: Designing a pediatric severe sepsis screening tool. Front Pediatr 2014; 2:56

41. Balamuth F, Alpern ER, Abbadessa MK, et al: Improving recognition of pediatric severe sepsis in the emergency department: Contributions of a vital sign-based electronic alert and bedside clinician identifica-tion. Ann Emerg Med 2017; 70:759–768.e2

42. Bradshaw C, Goodman I, Rosenberg R, et al: Implementation of an inpatient pediatric sepsis identification pathway. Pediatrics 2016; 137:e20144082

43. Balamuth F, Alpern ER, Grundmeier RW, et al: Comparison of two sepsis recognition methods in a pediatric emergency department. Acad Emerg Med 2015; 22:1298–1306

44. Schlapbach LJ, Weiss SL, Wolf J: Reducing collateral damage from mandates for time to antibiotics in pediatric sepsis-primum non nocere. JAMA Pediatr 2019; 173:409–410

45. Leigh S, Grant A, Murray N, et al: The cost of diagnostic uncertainty: A prospective economic analysis of febrile children attending an NHS emergency department. BMC Med 2019; 17:48

46. Duncan H, Hutchison J, Parshuram CS: The pediatric early warning system score: A severity of illness score to predict urgent medical need in hospitalized children. J Crit Care 2006; 21:271–278

47. Chapman SM, Wray J, Oulton K, et al: ‘The Score Matters’: Wide variations in predictive performance of 18 paediatric track and trigger systems. Arch Dis Child 2017; 102:487–495

48. Lambert V, Matthews A, MacDonell R, et al: Paediatric early warning systems for detecting and responding to clinical deterioration in chil-dren: A systematic review. BMJ Open 2017; 7:e014497

49. Hernandez G, Bellomo R, Bakker J: The ten pitfalls of lactate clear-ance in sepsis. Intensive Care Med 2019; 45:82–85

50. Rhodes A, Evans LE, Alhazzani W, et al: Surviving sepsis campaign: International guidelines for management of sepsis and septic shock: 2016. Intensive Care Med 2017; 43:304–377

51. Levy MM, Evans LE, Rhodes A: The surviving sepsis campaign bundle: 2018 update. Intensive Care Med 2018; 44:925–928

52. Bai Z, Zhu X, Li M, et al: Effectiveness of predicting in-hospital mor-tality in critically ill children by assessing blood lactate levels at admis-sion. BMC Pediatr 2014; 14:83

53. Chen M, Lu X, Hu L, et al: Development and validation of a mor-tality risk model for pediatric sepsis. Medicine (Baltimore) 2017; 96:e6923

54. Scott HF, Brou L, Deakyne SJ, et al: Association between early lac-tate levels and 30-day mortality in clinically suspected sepsis in chil-dren. JAMA Pediatr 2017; 171:249–255

55. Schlapbach LJ, MacLaren G, Straney L: Venous vs arterial lactate and 30-day mortality in pediatric sepsis. JAMA Pediatr 2017; 171:813

56. Scott HF, Brou L, Deakyne SJ, et al: Lactate clearance and normali-zation and prolonged organ dysfunction in pediatric sepsis. J Pediatr 2016; 170:149–155.e1

57. Kortz TB, Axelrod DM, Chisti MJ, et al: Clinical outcomes and mortality before and after implementation of a pediatric sepsis protocol in a limited resource setting: A retrospective cohort study in Bangladesh. PLoS One 2017; 12:e0181160

58. Long E, Babl FE, Angley E, et al: A prospective quality improvement study in the emergency department targeting paediatric sepsis. Arch Dis Child 2016; 101:945–950

59. Workman JK, Ames SG, Reeder RW, et al: Treatment of pediatric septic shock with the surviving sepsis campaign guidelines and PICU patient outcomes. Pediatr Crit Care Med 2016; 17:e451–e458

60. Larsen GY, Mecham N, Greenberg R: An emergency department septic shock protocol and care guideline for children initiated at tri-age. Pediatrics 2011; 127:e1585–e1592

61. Tuuri RE, Gehrig MG, Busch CE, et al: “Beat the Shock Clock”: An interprofessional team improves pediatric septic shock care. Clin Pediatr (Phila) 2016; 55:626–638

62. McMullan BJ, Bowen A, Blyth CC, et al: Epidemiology and mortality of Staphylococcus aureus bacteremia in Australian and New Zealand children. JAMA Pediatr 2016; 170:979–986

63. Weiss SL, Fitzgerald JC, Balamuth F, et al: Delayed antimicrobial therapy increases mortality and organ dysfunction duration in pedi-atric sepsis. Crit Care Med 2014; 42:2409–2417

64. Martinón-Torres F, Salas A, Rivero-Calle I, et al; EUCLIDS Consortium: Life-threatening infections in children in Europe (the EUCLIDS pro-ject): A prospective cohort study. Lancet Child Adolesc Health 2018; 2:404–414

65. Agyeman PKA, Schlapbach LJ, Giannoni E, et al; Swiss Pediatric Sepsis Study: Epidemiology of blood culture-proven bacterial sepsis in children in Switzerland: A population-based cohort study. Lancet Child Adolesc Health 2017; 1:124–133

66. Lucignano B, Ranno S, Liesenfeld O, et al: Multiplex PCR allows rapid and accurate diagnosis of bloodstream infections in new-borns and children with suspected sepsis. J Clin Microbiol 2011; 49:2252–2258

67. Rhee C, Filbin MR, Massaro AF, et al; Centers for Disease Control and Prevention (CDC) Prevention Epicenters Program: Compliance with the national SEP-1 quality measure and association with sepsis outcomes: A multicenter retrospective cohort study. Crit Care Med 2018; 46:1585–1591

68. Kumar A, Roberts D, Wood KE, et al: Duration of hypotension before initiation of effective antimicrobial therapy is the critical determinant of survival in human septic shock. Crit Care Med 2006; 34:1589–1596

69. Ferrer R, Martin-Loeches I, Phillips G, et al: Empiric antibiotic treat-ment reduces mortality in severe sepsis and septic shock from the first hour: Results from a guideline-based performance improvement program. Crit Care Med 2014; 42:1749–1755

70. Barie PS, Hydo LJ, Shou J, et al: Influence of antibiotic therapy on mortality of critical surgical illness caused or complicated by infection. Surg Infect (Larchmt) 2005; 6:41–54

71. Barochia AV, Cui X, Vitberg D, et al: Bundled care for septic shock: An analysis of clinical trials. Crit Care Med 2010; 38:668–678

72. Gaieski DF, Mikkelsen ME, Band RA, et al: Impact of time to antibiot-ics on survival in patients with severe sepsis or septic shock in whom early goal-directed therapy was initiated in the emergency depart-ment. Crit Care Med 2010; 38:1045–1053

73. Klompas M, Calandra T, Singer M: Antibiotics for sepsis-finding the equilibrium. JAMA 2018; 320:1433–1434

74. Mi MY, Klompas M, Evans L: Early administration of antibiotics for suspected sepsis. N Engl J Med 2019; 380:593–596

75. Kumar A, Ellis P, Arabi Y, et al; Cooperative Antimicrobial Therapy of Septic Shock Database Research Group: Initiation of inappro-priate antimicrobial therapy results in a fivefold reduction of survival in human septic shock. Chest 2009; 136:1237–1248

76. Kimberlin D, Brady M, Jackson M, et al: American Academy of Pediatrics Committee on Infectious Diseases Red Book® 2018. 2018. Available at: https://redbook.solutions.aap.org/Book.aspx?bookid=2205. Accessed August 18, 2019

77. Sharland M, Butler K, Cant A, et al: European Society for Paediatric Infectious Diseases Manual of Childhood Infections -The Blue Book. 2016. Available at: https://global.oup.com/academic/product/manual-of-childhood-infections-9780198729228?cc=gb&lang=en&. Accessed August 18, 2019

78. Lehrnbecher T, Robinson P, Fisher B, et al: Guideline for the manage-ment of fever and neutropenia in children with cancer and hematopoi-etic stem-cell transplantation recipients: 2017 update. J Clin Oncol 2017; 35:2082–2094

Page 40: Surviving Sepsis Campaign International Guidelines for the ... · In 2001, the Surviving Sepsis Campaign (SSC) was formed by the Society of Critical Care Medicine (SCCM), European

Copyright © 2020 by the Society of Critical Care Medicine and the World Federation of Pediatric Intensive and Critical Care Societies.Unauthorized reproduction of this article is prohibited

Online Special Article

Pediatric Critical Care Medicine www.pccmjournal.org e91

79. Iroh Tam PY, Musicha P, Kawaza K, et al: Emerging resistance to em-piric antimicrobial regimens for pediatric bloodstream infections in Malawi (1998-2017). Clin Infect Dis 2019; 69:61–68

80. Malosh RE, Martin ET, Heikkinen T, et al: Efficacy and safety of osel-tamivir in children: Systematic review and individual patient data meta-analysis of randomized controlled trials. Clin Infect Dis 2018; 66:1492–1500

81. Kumar A: Early versus late oseltamivir treatment in severely ill patients with 2009 pandemic influenza A (H1N1): Speed is life. J Antimicrob Chemother 2011; 66:959–963

82. Goodman KE, Lessler J, Cosgrove SE, et al; Antibacterial Resistance Leadership Group: A clinical decision tree to predict whether a bac-teremic patient is infected with an extended-spectrum β-lactamase-producing organism. Clin Infect Dis 2016; 63:896–903

83. Carapetis JR, Jacoby P, Carville K, et al: Effectiveness of clindamycin and intravenous immunoglobulin, and risk of disease in contacts, in invasive group a streptococcal infections. Clin Infect Dis 2014; 59:358–365

84. Guo Y, Gao W, Yang H, et al: De-escalation of empiric antibiotics in patients with severe sepsis or septic shock: A meta-analysis. Heart Lung 2016; 45:454–459

85. Hamdy RF, Zaoutis TE, Seo SK: Antifungal stewardship consider-ations for adults and pediatrics. Virulence 2017; 8:658–672

86. Dierig A, Berger C, Agyeman PKA, et al; Swiss Pediatric Sepsis Study: Time-to-positivity of blood cultures in children with sepsis. Front Pediatr 2018; 6:222

87. Lin GL, McGinley JP, Drysdale SB, et al: Epidemiology and immune pathogenesis of viral sepsis. Front Immunol 2018; 9:2147

88. Paul M, Dickstein Y, Schlesinger A, et al: Beta-lactam versus beta-lactam-aminoglycoside combination therapy in cancer patients with neutropenia. Cochrane Database Syst Rev 2013; (6):CD003038

89. Paul M, Lador A, Grozinsky-Glasberg S, et al: Beta lactam antibiotic monotherapy versus beta lactam-aminoglycoside antibiotic com-bination therapy for sepsis. Cochrane Database Syst Rev 2014; (1):CD003344

90. Sjövall F, Perner A, Hylander Møller M: Empirical mono- versus com-bination antibiotic therapy in adult intensive care patients with severe sepsis - a systematic review with meta-analysis and trial sequential analysis. J Infect 2017; 74:331–344

91. Baqui AH, Saha SK, Ahmed AS, et al; Projahnmo Study Group in Bangladesh: Safety and efficacy of alternative antibiotic regimens compared with 7 day injectable procaine benzylpenicillin and gen-tamicin for outpatient treatment of neonates and young infants with clinical signs of severe infection when referral is not possible: A ran-domised, open-label, equivalence trial. Lancet Glob Health 2015; 3:e279–e287

92. Metsvaht T, Ilmoja ML, Parm Ü, et al: Comparison of ampicillin plus gentamicin vs. penicillin plus gentamicin in empiric treatment of neo-nates at risk of early onset sepsis. Acta Paediatr 2010; 99:665–672

93. Pasha YZ, Ahmadpour-Kacho M, Behmadi R, et al: 3-day versus 5-day course of intravenous antibiotics for suspected early onset neonatal sepsis: A randomized controlled trial. Iran J Pediatr 2014; 24:673–678

94. Ramasamy S, Biswal N, Bethou A, et al: Comparison of two empiric antibiotic regimen in late onset neonatal sepsis–a randomized con-trolled trial. J Trop Pediatr 2014; 60:83–86

95. Taheri PA, Eslamieh H, Salamati P: Is ceftizoxime an appropriate sur-rogate for amikacin in neonatal sepsis treatment? A randomized clin-ical trial. Acta Med Iran 2011; 49:499–503

96. Tewari VV, Jain N: Monotherapy with amikacin or piperacillin-tazo-bactum empirically in neonates at risk for early-onset sepsis: A ran-domized controlled trial. J Trop Pediatr 2014; 60:297–302

97. Tshefu A, Lokangaka A, Ngaima S, et al; African Neonatal Sepsis Trial (AFRINEST) group: Simplified antibiotic regimens compared with injectable procaine benzylpenicillin plus gentamicin for treat-ment of neonates and young infants with clinical signs of possible se-rious bacterial infection when referral is not possible: A randomised, open-label, equivalence trial. Lancet 2015; 385:1767–1776

98. Zaidi AK, Tikmani SS, Warraich HJ, et al: Community-based treat-ment of serious bacterial infections in newborns and young infants:

A randomized controlled trial assessing three antibiotic regimens. Pediatr Infect Dis J 2012; 31:667–672

99. Ibrahim SL, Zhang L, Brady TM, et al: Low-dose gentamicin for un-complicated enterococcus faecalis bacteremia may be nephrotoxic in children. Clin Infect Dis 2015; 61:1119–1124

100. Tamma PD, Turnbull AE, Harris AD, et al: Less is more: Combination antibiotic therapy for the treatment of gram-negative bacteremia in pediatric patients. JAMA Pediatr 2013; 167:903–910

101. Falagas ME, Lourida P, Poulikakos P, et al: Antibiotic treatment of infections due to carbapenem-resistant Enterobacteriaceae: Systematic evaluation of the available evidence. Antimicrob Agents Chemother 2014; 58:654–663

102. Bass SN, Bauer SR, Neuner EA, et al: Impact of combination antimi-crobial therapy on mortality risk for critically ill patients with carbape-nem-resistant bacteremia. Antimicrob Agents Chemother 2015; 59:3748–3753

103. Chiotos K, Tamma PD, Flett KB, et al: Increased 30-day mortality as-sociated with carbapenem-resistant Enterobacteriaceae in children. Open Forum Infect Dis 2018; 5:ofy222

104. Baltimore RS, Gewitz M, Baddour LM, et al; American Heart Association Rheumatic Fever, Endocarditis, and Kawasaki Disease Committee of the Council on Cardiovascular Disease in the Young and the Council on Cardiovascular and Stroke Nursing: Infective en-docarditis in childhood: 2015 update: A scientific statement from the American Heart Association. Circulation 2015; 132:1487–1515

105. Hanretty AM, Kaur I, Evangelista AT, et al: Pharmacokinetics of the meropenem component of meropenem-vaborbactam in the treat-ment of KPC-producing Klebsiella pneumoniae bloodstream infec-tion in a pediatric patient. Pharmacotherapy 2018; 38:e87–e91

106. Costa Pde O, Atta EH, Silva AR: Infection with multidrug-resistant gram-negative bacteria in a pediatric oncology intensive care unit: Risk factors and outcomes. J Pediatr (Rio J) 2015; 91:435–441

107. Shankar K, Radhakrishnan V, Vijayakumar V, et al: Prevalence of multi-drug resistant organisms in stool of paediatric patients with acute leukaemia and correlation with blood culture positivity: A single institution experience. Pediatr Blood Cancer 2018; 65. Epub 2017 Aug 2.

108. Pereira CA, Petrilli AS, Carlesse FA, et al: Cefepime monotherapy is as effective as ceftriaxone plus amikacin in pediatric patients with cancer and high-risk febrile neutropenia in a randomized compar-ison. J Microbiol Immunol Infect 2009; 42:141–147

109. Petrilli AS, Cypriano M, Dantas LS, et al: Evaluation of ticarcillin/clavulanic acid versus ceftriaxone plus amikacin for fever and neutro-penia in pediatric patients with leukemia and lymphoma. Braz J Infect Dis 2003; 7:111–120

110. Daneman N, Low DE, McGeer A, et al: At the threshold: Defining clin-ically meaningful resistance thresholds for antibiotic choice in com-munity-acquired pneumonia. Clin Infect Dis 2008; 46:1131–1138

111. Mandell LA, Wunderink RG, Anzueto A, et al; Infectious Diseases Society of America; American Thoracic Society: Infectious Diseases Society of America/American Thoracic Society consensus guide-lines on the management of community-acquired pneumonia in adults. Clin Infect Dis 2007; 44(Suppl 2):S27–S72

112. Liu C, Bayer A, Cosgrove SE, et al; Infectious Diseases Society of America: Clinical practice guidelines by the Infectious Diseases Society of America for the treatment of methicillin-resistant Staphylococcus aureus infections in adults and children. Clin Infect Dis 2011; 52:e18–e55

113. Cies JJ, Moore WS 2nd, Enache A, et al: β-lactam therapeutic drug management in the PICU. Crit Care Med 2018; 46:272–279

114. Nehus EJ, Mizuno T, Cox S, et al: Pharmacokinetics of meropenem in children receiving continuous renal replacement therapy: Validation of clinical trial simulations. J Clin Pharmacol 2016; 56:291–297

115. Guilhaumou R, Benaboud S, Bennis Y, et al: Optimization of the treatment with beta-lactam antibiotics in critically ill patients-guide-lines from the French Society of Pharmacology and Therapeutics (Société Française de Pharmacologie et Thérapeutique-SFPT) and the French Society of Anaesthesia and Intensive Care Medicine (Société Française d’Anesthésie et Réanimation-SFAR). Crit Care 2019; 23:104

Page 41: Surviving Sepsis Campaign International Guidelines for the ... · In 2001, the Surviving Sepsis Campaign (SSC) was formed by the Society of Critical Care Medicine (SCCM), European

Copyright © 2020 by the Society of Critical Care Medicine and the World Federation of Pediatric Intensive and Critical Care Societies.Unauthorized reproduction of this article is prohibited

Copyright © 2020 by the Society of Critical Care Medicine and the World Federation of Pediatric Intensive and Critical Care Societies.Unauthorized reproduction of this article is prohibited

Weiss et al

e92 www.pccmjournal.org February 2020 • Volume 21 • Number 2

116. Roberts JA, Abdul-Aziz MH, Lipman J, et al; International Society of Anti-Infective Pharmacology and the Pharmacokinetics and Pharmacodynamics Study Group of the European Society of Clinical Microbiology and Infectious Diseases: Individualised antibiotic dos-ing for patients who are critically ill: Challenges and potential solu-tions. Lancet Infect Dis 2014; 14:498–509

117. Udy AA, Varghese JM, Altukroni M, et al: Subtherapeutic initial β-lactam concentrations in select critically ill patients: Association between augmented renal clearance and low trough drug concentra-tions. Chest 2012; 142:30–39

118. Cotta MO, Roberts JA, Lipman J: Antibiotic dose optimization in crit-ically ill patients. Med Intensiva 2015; 39:563–572

119. De Bleecker JL, Meire VI, Declercq W, et al: Immunolocalization of tumor necrosis factor-alpha and its receptors in inflammatory myopa-thies. Neuromuscul Disord 1999; 9:239–246

120. Van Der Heggen T, Dhont E, Peperstraete H, et al: Augmented renal clearance: A common condition in critically ill children. Pediatr Nephrol 2019; 34:1099–1106

121. Rizk NA, Kanafani ZA, Tabaja HZ, et al: Extended infusion of beta-lactam antibiotics: Optimizing therapy in critically-ill patients in the era of antimicrobial resistance. Expert Rev Anti Infect Ther 2017; 15:645–652

122. Demirjian A, Finkelstein Y, Nava-Ocampo A, et al: A randomized con-trolled trial of a vancomycin loading dose in children. Pediatr Infect Dis J 2013; 32:1217–1223

123. Godbout EJ, Pakyz AL, Markley JD, et al: Pediatric antimicrobial stewardship: State of the art. Curr Infect Dis Rep 2018; 20:39

124. Weiss CH, Persell SD, Wunderink RG, et al: Empiric antibiotic, me-chanical ventilation, and central venous catheter duration as poten-tial factors mediating the effect of a checklist prompting intervention on mortality: An exploratory analysis. BMC Health Serv Res 2012; 12:198

125. Weiss CH, Moazed F, McEvoy CA, et al: Prompting physicians to address a daily checklist and process of care and clinical outcomes: A single-site study. Am J Respir Crit Care Med 2011; 184:680–686

126. Schuetz P, Briel M, Christ-Crain M, et al: Procalcitonin to guide ini-tiation and duration of antibiotic treatment in acute respiratory infec-tions: An individual patient data meta-analysis. Clin Infect Dis 2012; 55:651–662

127. Matthaiou DK, Ntani G, Kontogiorgi M, et al: An ESICM systematic review and meta-analysis of procalcitonin-guided antibiotic therapy algorithms in adult critically ill patients. Intensive Care Med 2012; 38:940–949

128. Prkno A, Wacker C, Brunkhorst FM, et al: Procalcitonin-guided therapy in intensive care unit patients with severe sepsis and septic shock–a systematic review and meta-analysis. Crit Care 2013; 17:R291

129. Westwood M, Ramaekers B, Whiting P, et al: Procalcitonin testing to guide antibiotic therapy for the treatment of sepsis in intensive care settings and for suspected bacterial infection in emergency de-partment settings: A systematic review and cost-effectiveness anal-ysis. Health Technol Assess 2015; 19:v–xxv, 1–236

130. Wacker C, Prkno A, Brunkhorst FM, et al: Procalcitonin as a diag-nostic marker for sepsis: A systematic review and meta-analysis. Lancet Infect Dis 2013; 13:426–435

131. de Jong E, van Oers JA, Beishuizen A, et al: Efficacy and safety of procalcitonin guidance in reducing the duration of antibiotic treat-ment in critically ill patients: A randomised, controlled, open-label trial. Lancet Infect Dis 2016; 16:819–827

132. Stocker M, van Herk W, El Helou S, et al; NeoPInS Study Group: Procalcitonin-guided decision making for duration of antibiotic therapy in neonates with suspected early-onset sepsis: A multicentre, randomised controlled trial (NeoPIns). Lancet 2017; 390:871–881

133. Public Health England: Start Smart - Then Focus. United Kingdom, Public Health England, 2015. Available at: https://www.gov.uk/government/publications/antimicrobial-stewardship-start-smart-then-focus#history. Accessed August 18, 2019

134. Bamberger DM: Outcome of medical treatment of bacterial abscesses without therapeutic drainage: Review of cases reported in the literature. Clin Infect Dis 1996; 23:592–603

135. Osmon DR, Berbari EF, Berendt AR, et al; Infectious Diseases Society of America: Diagnosis and management of prosthetic joint infection: Clinical practice guidelines by the Infectious Diseases Society of America. Clin Infect Dis 2013; 56:e1–e25

136. Baddour LM, Wilson WR, Bayer AS, et al; American Heart Association Committee on Rheumatic Fever, Endocarditis, and Kawasaki Disease of the Council on Cardiovascular Disease in the Young, Council on Clinical Cardiology, Council on Cardiovascular Surgery and Anesthesia, and Stroke Council: Infective endocarditis in adults: Diagnosis, antimicrobial therapy, and management of com-plications: A scientific statement for healthcare professionals from the American Heart Association. Circulation 2015; 132:1435–1486

137. Yahav D, Franceschini E, Koppel F, et al; Bacteremia Duration Study Group: Seven versus 14 days of antibiotic therapy for uncompli-cated gram-negative bacteremia: A noninferiority randomized con-trolled trial. Clin Infect Dis 2019; 69:1091–1098

138. Chotiprasitsakul D, Han JH, Cosgrove SE, et al; Antibacterial Resistance Leadership Group: Comparing the outcomes of adults with Enterobacteriaceae bacteremia receiving short-course versus prolonged-course antibiotic therapy in a multicenter, propensity score-matched cohort. Clin Infect Dis 2018; 66:172–177

139. Chong YP, Moon SM, Bang KM, et al: Treatment duration for un-complicated Staphylococcus aureus bacteremia to prevent relapse: Analysis of a prospective observational cohort study. Antimicrob Agents Chemother 2013; 57:1150–1156

140. Chowdhary G, Dutta S, Narang A: Randomized controlled trial of 7-day vs. 14-day antibiotics for neonatal sepsis. J Trop Pediatr 2006; 52:427–432

141. Liu C, Bayer A, Cosgrove SE, et al: Clinical practice guidelines by the Infectious Diseases Society of America for the treatment of methicillin-resistant Staphylococcus aureus infections in adults and children: Executive summary. Clin Infect Dis 2011; 52:285–292

142. Berrevoets MAH, Kouijzer IJE, Aarntzen EHJG, et al: 18F-FDG PET/CT optimizes treatment in Staphylococcus Aureus bacteremia and is associated with reduced mortality. J Nucl Med 2017; 58:1504–1510

143. McMullan BJ, Andresen D, Blyth CC, et al; ANZPID-ASAP group: Antibiotic duration and timing of the switch from intravenous to oral route for bacterial infections in children: Systematic review and guidelines. Lancet Infect Dis 2016; 16:e139–e152

144. Alexander VN, Northrup V, Bizzarro MJ: Antibiotic exposure in the newborn intensive care unit and the risk of necrotizing enterocolitis. J Pediatr 2011; 159:392–397

145. Paganini H, Rodriguez Brieshcke T, Santos P, et al: Risk factors for nosocomial candidaemia: A case-control study in children. J Hosp Infect 2002; 50:304–308

146. Zaoutis TE, Prasad PA, Localio AR, et al: Risk factors and predictors for candidemia in pediatric intensive care unit patients: Implications for prevention. Clin Infect Dis 2010; 51:e38–e45

147. Chatterjee A, Modarai M, Naylor NR, et al: Quantifying drivers of an-tibiotic resistance in humans: A systematic review. Lancet Infect Dis 2018; 18:e368–e378

148. Brown KA, Khanafer N, Daneman N, et al: Meta-analysis of antibiot-ics and the risk of community-associated Clostridium difficile infec-tion. Antimicrob Agents Chemother 2013; 57:2326–2332

149. Chuang YY, Huang YC, Lee CY, et al: Methicillin-resistant Staphylococcus aureus bacteraemia in neonatal intensive care units: An analysis of 90 episodes. Acta Paediatr 2004; 93:786–790

150. Couto RC, Barbosa JA, Pedrosa TM, et al: C-reactive protein-guided approach may shorten length of antimicrobial treatment of culture-proven late-onset sepsis: An intervention study. Braz J Infect Dis 2007; 11:240–245

151. Dimopoulos G, Matthaiou DK, Karageorgopoulos DE, et al: Short- versus long-course antibacterial therapy for community-acquired pneumonia: A meta-analysis. Drugs 2008; 68:1841–1854

152. Havey TC, Fowler RA, Daneman N: Duration of antibiotic therapy for bacteremia: A systematic review and meta-analysis. Crit Care 2011; 15:R267

153. Hemels MA, van den Hoogen A, Verboon-Maciolek MA, et al: Shortening the antibiotic course for the treatment of neonatal coagulase-negative staphylococcal sepsis: Fine with three days? Neonatology 2012; 101:101–105

Page 42: Surviving Sepsis Campaign International Guidelines for the ... · In 2001, the Surviving Sepsis Campaign (SSC) was formed by the Society of Critical Care Medicine (SCCM), European

Copyright © 2020 by the Society of Critical Care Medicine and the World Federation of Pediatric Intensive and Critical Care Societies.Unauthorized reproduction of this article is prohibited

Online Special Article

Pediatric Critical Care Medicine www.pccmjournal.org e93

154. Karageorgopoulos DE, Valkimadi PE, Kapaskelis A, et al: Short versus long duration of antibiotic therapy for bacterial meningitis: A meta-analysis of randomised controlled trials in children. Arch Dis Child 2009; 94:607–614

155. Kavaliotis J, Manios SG, Kansouzidou A, et al: Treatment of child-hood bacterial meningitis with ceftriaxone once daily: Open, pro-spective, randomized, comparative study of short-course versus standard-length therapy. Chemotherapy 1989; 35:296–303

156. Lassi ZS, Imdad A, Bhutta ZA: Short-course versus long-course in-travenous therapy with the same antibiotic for severe community-acquired pneumonia in children aged two months to 59 months. Cochrane Database Syst Rev 2017; 10:CD008032

157. Lin TY, Chrane DF, Nelson JD, et al: Seven days of ceftriaxone therapy is as effective as ten days’ treatment for bacterial meningitis. JAMA 1985; 253:3559–3563

158. Linder N, Lubin D, Hernandez A, et al: Duration of vancomycin treat-ment for coagulase-negative Staphylococcus sepsis in very low birth weight infants. Br J Clin Pharmacol 2013; 76:58–64

159. Martin E, Hohl P, Guggi T, et al: Short course single daily ceftriaxone monotherapy for acute bacterial meningitis in children: Results of a Swiss multicenter study. Part I: Clinical results. Infection 1990; 18:70–77

160. Molyneux E, Nizami SQ, Saha S, et al; CSF 5 Study Group: 5 versus 10 days of treatment with ceftriaxone for bacterial meningitis in chil-dren: A double-blind randomised equivalence study. Lancet 2011; 377:1837–1845

161. Onakpoya IJ, Walker AS, Tan PS, et al: Overview of systematic reviews assessing the evidence for shorter versus longer duration antibiotic treatment for bacterial infections in secondary care. PLoS One 2018; 13:e0194858

162. Park SH, Milstone AM, Diener-West M, et al: Short versus prolonged courses of antibiotic therapy for children with uncomplicated Gram-negative bacteraemia. J Antimicrob Chemother 2014; 69:779–785

163. Rohatgi S, Dewan P, Faridi MMA, et al: Seven versus 10 days antibi-otic therapy for culture-proven neonatal sepsis: A randomised con-trolled trial. J Paediatr Child Health 2017; 53:556–562

164. Roine I, Ledermann W, Foncea LM, et al: Randomized trial of four vs. seven days of ceftriaxone treatment for bacterial meningitis in children with rapid initial recovery. Pediatr Infect Dis J 2000; 19:219–222

165. Singhi P, Kaushal M, Singhi S, et al: Seven days vs. 10 days ceftriax-one therapy in bacterial meningitis. J Trop Pediatr 2002; 48:273–279

166. Tsai MH, Huang YC, Chiu CH, et al: Nontyphoidal Salmonella bac-teremia in previously healthy children: Analysis of 199 episodes. Pediatr Infect Dis J 2007; 26:909–913

167. Yen MH, Huang YC, Chiu CH, et al: Duration of antimicrobial therapy for non-typhoid Salmonella bacteremia in healthy children. J Microbiol Immunol Infect 2002; 35:94–98

168. Jernelius H, Zbornik J, Bauer CA: One or three weeks’ treatment of acute pyelonephritis? A double-blind comparison, using a fixed com-bination of pivampicillin plus pivmecillinam. Acta Med Scand 1988; 223:469–477

169. Pakistan Multicentre Amoxycillin Short Course Therapy (MASCOT) pneumonia study group: Clinical efficacy of 3 days versus 5 days of oral amoxicillin for treatment of childhood pneumonia: A multicentre double-blind trial. Lancet 2002; 360:835–841

170. Agarwal G, Awasthi S, Kabra SK, et al; ISCAP Study Group: Three day versus five day treatment with amoxicillin for non-severe pneu-monia in young children: A multicentre randomised controlled trial. BMJ 2004; 328:791

171. Lagunes L, Encina B, Ramirez-Estrada S: Current understanding in source control management in septic shock patients: A review. Ann Transl Med 2016; 4:330

172. Martínez ML, Ferrer R, Torrents E, et al; Edusepsis Study Group: Impact of source control in patients with severe sepsis and septic shock. Crit Care Med 2017; 45:11–19

173. Fustes-Morales A, Gutierrez-Castrellon P, Duran-Mckinster C, et al: Necrotizing fasciitis: Report of 39 pediatric cases. Arch Dermatol 2002; 138:893–899

174. Bingöl-Koloğlu M, Yildiz RV, Alper B, et al: Necrotizing fasciitis in children: Diagnostic and therapeutic aspects. J Pediatr Surg 2007; 42:1892–1897

175. Endorf FW, Garrison MM, Klein MB, et al: Characteristics, thera-pies, and outcome of children with necrotizing soft tissue infections. Pediatr Infect Dis J 2012; 31:221–223

176. Nazemi KJ, Buescher ES, Kelly RE Jr, et al: Central venous catheter removal versus in situ treatment in neonates with enterobacteriaceae bacteremia. Pediatrics 2003; 111:e269–e274

177. Vasudevan C, Oddie SJ, McGuire W: Early removal versus expectant management of central venous catheters in neonates with blood-stream infection. Cochrane Database Syst Rev 2016; 4:CD008436

178. Rodriguez D, Park BJ, Almirante B, et al; Barcelona Candidemia Project Study Group: Impact of early central venous catheter re-moval on outcome in patients with candidaemia. Clin Microbiol Infect 2007; 13:788–793

179. Smith PB, Benjamin DK Jr, Cotten CM, et al: Is an increased dwell time of a peripherally inserted catheter associated with an increased risk of bloodstream infection in infants? Infect Control Hosp Epidemiol 2008; 29:749–753

180. Santhanam I, Sangareddi S, Venkataraman S, et al: A prospec-tive randomized controlled study of two fluid regimens in the initial management of septic shock in the emergency department. Pediatr Emerg Care 2008; 24:647–655

181. Sankar J, Ismail J, Sankar MJ, et al: Fluid bolus over 15-20 versus 5-10 minutes each in the first hour of resuscitation in children with septic shock: A randomized controlled trial. Pediatr Crit Care Med 2017; 18:e435–e445

182. Inwald DP, Canter R, Woolfall K, et al; PERUKI (Paediatric Emergency Research in the UK and Ireland) and PICS SG (Paediatric Intensive Care Society Study Group): Restricted fluid bolus volume in early septic shock: Results of the fluids in shock pilot trial. Arch Dis Child 2019; 104:426–431

183. Maitland K, Kiguli S, Opoka RO, et al; FEAST Trial Group: Mortality after fluid bolus in African children with severe infection. N Engl J Med 2011; 364:2483–2495

184. Carcillo JA, Davis AL, Zaritsky A: Role of early fluid resuscitation in pediatric septic shock. JAMA 1991; 266:1242–1245

185. Han YY, Carcillo JA, Dragotta MA, et al: Early reversal of pediatric-neonatal septic shock by community physicians is associated with improved outcome. Pediatrics 2003; 112:793–799

186. Carcillo JA, Kuch BA, Han YY, et al: Mortality and functional mor-bidity after use of PALS/APLS by community physicians. Pediatrics 2009; 124:500–508

187. van Paridon BM, Sheppard C, Garcia GG, et al: Timing of antibiot-ics, volume, and vasoactive infusions in children with sepsis admitted to intensive care. Crit Care 2015; 19:293

188. Houston KA, George EC, Maitland K: Implications for paediatric shock management in resource-limited settings: A perspective from the FEAST trial. Crit Care 2018; 22:119

189. Voigt J, Waltzman M, Lottenberg L: Intraosseous vascular access for in-hospital emergency use: A systematic clinical review of the litera-ture and analysis. Pediatr Emerg Care 2012; 28:185–199

190. de Oliveira CF, de Oliveira DS, Gottschald AF, et al: ACCM/PALS haemodynamic support guidelines for paediatric septic shock: An outcomes comparison with and without monitoring central venous oxygen saturation. Intensive Care Med 2008; 34:1065–1075

191. Arya B, Kerstein D, Leu CS, et al: Echocardiographic assessment of right atrial pressure in a pediatric and young adult population. Pediatr Cardiol 2016; 37:558–567

192. Semler MW, Self WH, Wanderer JP, et al; SMART Investigators and the Pragmatic Critical Care Research Group: Balanced crystalloids versus saline in critically ill adults. N Engl J Med 2018; 378:829–839

193. Weiss SL, Keele L, Balamuth F, et al: Crystalloid fluid choice and clinical outcomes in pediatric sepsis: A matched retrospective co-hort study. J Pediatr 2017; 182:304–310.e10

194. Emrath ET, Fortenberry JD, Travers C, et al: Resuscitation with bal-anced fluids is associated with improved survival in pediatric severe sepsis. Crit Care Med 2017; 45:1177–1183

Page 43: Surviving Sepsis Campaign International Guidelines for the ... · In 2001, the Surviving Sepsis Campaign (SSC) was formed by the Society of Critical Care Medicine (SCCM), European

Copyright © 2020 by the Society of Critical Care Medicine and the World Federation of Pediatric Intensive and Critical Care Societies.Unauthorized reproduction of this article is prohibited

Copyright © 2020 by the Society of Critical Care Medicine and the World Federation of Pediatric Intensive and Critical Care Societies.Unauthorized reproduction of this article is prohibited

Weiss et al

e94 www.pccmjournal.org February 2020 • Volume 21 • Number 2

195. Self WH, Semler MW, Wanderer JP, et al; SALT-ED Investigators: Balanced crystalloids versus saline in noncritically ill adults. N Engl J Med 2018; 378:819–828

196. Perner A, Haase N, Guttormsen AB, et al; 6S Trial Group; Scandinavian Critical Care Trials Group: Hydroxyethyl starch 130/0.42 versus ringer’s acetate in severe sepsis. N Engl J Med 2012; 367:124–134

197. Myburgh JA, Finfer S, Bellomo R, et al; CHEST Investigators; Australian and New Zealand Intensive Care Society Clinical Trials Group: Hydroxyethyl starch or saline for fluid resuscitation in inten-sive care. N Engl J Med 2012; 367:1901–1911

198. Zarychanski R, Abou-Setta AM, Turgeon AF, et al: Association of hydroxyethyl starch administration with mortality and acute kidney injury in critically ill patients requiring volume resuscitation: A system-atic review and meta-analysis. JAMA 2013; 309:678–688

199. McIntyre L: Hydroxyethyl Starch for Fluid Resuscitation. 2012. Available at: https://criticalcarecanada.com/presentations/2012/hydroxyethyl_starch_for_fluid_resuscitation.pdf. Accessed August 18, 2019

200. European Medicines Agency: Hydroxyethyl-Starch Solutions for Infusion to Be Suspended – CMDh Endorses PRAC Recommendation. 2018. Available at: https://www.ema.europa.eu/en/news/hydroxyethyl-starch-solutions-infusion-be-suspended-cmdh-endorses-prac-recommendation. Accessed August 18, 2019

201. Upadhyay M, Singhi S, Murlidharan J, et al: Randomized evaluation of fluid resuscitation with crystalloid (saline) and colloid (polymer from degraded gelatin in saline) in pediatric septic shock. Indian Pediatr 2005; 42:223–231

202. Lamontagne F, Day AG, Meade MO, et al: Pooled analysis of higher versus lower blood pressure targets for vasopressor therapy septic and vasodilatory shock. Intensive Care Med 2018; 44:12–21

203. Davis AL, Carcillo JA, Aneja RK, et al: American College of Critical Care Medicine clinical practice parameters for hemodynamic sup-port of pediatric and neonatal septic shock. Crit Care Med 2017; 45:1061–1093

204. Tibby SM, Hatherill M, Marsh MJ, et al: Clinicians’ abilities to esti-mate cardiac index in ventilated children and infants. Arch Dis Child 1997; 77:516–518

205. Brierley J, Peters MJ: Distinct hemodynamic patterns of septic shock at presentation to pediatric intensive care. Pediatrics 2008; 122:752–759

206. Egan JR, Festa M, Cole AD, et al: Clinical assessment of cardiac per-formance in infants and children following cardiac surgery. Intensive Care Med 2005; 31:568–573

207. Ranjit S, Kissoon N: Bedside echocardiography is useful in assess-ing children with fluid and inotrope resistant septic shock. Indian J Crit Care Med 2013; 17:224–230

208. Ranjit S, Aram G, Kissoon N, et al: Multimodal monitoring for he-modynamic categorization and management of pediatric septic shock: A pilot observational study*. Pediatr Crit Care Med 2014; 15:e17–e26

209. Razavi A, Newth CJL, Khemani RG, et al: Cardiac output and sys-temic vascular resistance: Clinical assessment compared with a noninvasive objective measurement in children with shock. J Crit Care 2017; 39:6–10

210. El-Nawawy AA, Abdelmohsen AM, Hassouna HM: Role of echocar-diography in reducing shock reversal time in pediatric septic shock: A randomized controlled trial. J Pediatr (Rio J) 2018; 94:31–39

211. Gan H, Cannesson M, Chandler JR, et al: Predicting fluid respon-siveness in children: A systematic review. Anesth Analg 2013; 117:1380–1392

212. Rivers E, Nguyen B, Havstad S, et al; Early Goal-Directed Therapy Collaborative Group: Early goal-directed therapy in the treat-ment of severe sepsis and septic shock. N Engl J Med 2001; 345:1368–1377

213. Peake SL, Delaney A, Bailey M, et al; ARISE Investigators, ANZICS Clinical Trials Group: Goal-directed resuscitation for patients with early septic shock. N Engl J Med 2014; 371:1496–1506

214. Yealy DM, Kellum JA, Huang DT, et al; ProCESS Investigators: A ran-domized trial of protocol-based care for early septic shock. N Engl J Med 2014; 370:1683–1693

215. Mouncey PR, Osborn TM, Power GS, et al; ProMISe Trial Investigators: Trial of early, goal-directed resuscitation for septic shock. N Engl J Med 2015; 372:1301–1311

216. Jansen TC, van Bommel J, Schoonderbeek FJ, et al; LACTATE study group: Early lactate-guided therapy in intensive care unit patients: A multicenter, open-label, randomized controlled trial. Am J Respir Crit Care Med 2010; 182:752–761

217. Jones AE, Shapiro NI, Trzeciak S, et al; Emergency Medicine Shock Research Network (EMShockNet) Investigators: Lactate clearance vs central venous oxygen saturation as goals of early sepsis therapy: A randomized clinical trial. JAMA 2010; 303:739–746

218. Tian HH, Han SS, Lv CJ, et al: [The effect of early goal lactate clearance rate on the outcome of septic shock patients with se-vere pneumonia]. Zhongguo Wei Zhong Bing Ji Jiu Yi Xue 2012; 24:42–45

219. Yu B, Tian HY, Hu ZJ, et al: [Comparison of the effect of fluid re-suscitation as guided either by lactate clearance rate or by central venous oxygen saturation in patients with sepsis]. Zhonghua Wei Zhong Bing Ji Jiu Yi Xue 2013; 25:578–583

220. Lyu X, Xu Q, Cai G, et al: [Efficacies of fluid resuscitation as guided by lactate clearance rate and central venous oxygen satura-tion in patients with septic shock]. Zhonghua Yi Xue Za Zhi 2015; 95:496–500

221. Zhou X, Liu D, Su L, et al: Use of stepwise lactate kinetics-oriented hemodynamic therapy could improve the clinical outcomes of patients with sepsis-associated hyperlactatemia. Crit Care 2017; 21:33

222. Ventura AM, Shieh HH, Bousso A, et al: Double-blind prospective randomized controlled trial of dopamine versus epinephrine as first-line vasoactive drugs in pediatric septic shock. Crit Care Med 2015; 43:2292–2302

223. Ramaswamy KN, Singhi S, Jayashree M, et al: Double-blind random-ized clinical trial comparing dopamine and epinephrine in pediatric fluid-refractory hypotensive septic shock. Pediatr Crit Care Med 2016; 17:e502–e512

224. Piva J, Alquati T, Garcia PC, et al: Norepinephrine infusion increases urine output in children under sedative and analgesic infusion. Rev Assoc Med Bras (1992) 2014; 60:208–215

225. Avni T, Lador A, Lev S, et al: Vasopressors for the treatment of septic shock: Systematic review and meta-analysis. PLoS One 2015; 10:e0129305

226. Arikan AA, Zappitelli M, Goldstein SL, et al: Fluid overload is associ-ated with impaired oxygenation and morbidity in critically ill children. Pediatr Crit Care Med 2012; 13:253–258

227. Lampin ME, Rousseaux J, Botte A, et al: Noradrenaline use for septic shock in children: Doses, routes of administration and complications. Acta Paediatr 2012; 101:e426–e430

228. Patregnani JT, Sochet AA, Klugman D: Short-term peripheral vaso-active infusions in pediatrics: Where is the harm? Pediatr Crit Care Med 2017; 18:e378–e381

229. Choong K, Bohn D, Fraser DD, et al; Canadian Critical Care Trials Group: Vasopressin in pediatric vasodilatory shock: A multicenter randomized controlled trial. Am J Respir Crit Care Med 2009; 180:632–639

230. Baldasso E, Ramos Garcia PC, Piva JP, et al: Hemodynamic and metabolic effects of vasopressin infusion in children with shock. J Pediatr (Rio J) 2007; 83:S137–S145

231. Yildizdas D, Yapicioglu H, Celik U, et al: Terlipressin as a rescue therapy for catecholamine-resistant septic shock in children. Intensive Care Med 2008; 34:511–517

232. McIntyre WF, Um KJ, Alhazzani W, et al: Association of vasopressin plus catecholamine vasopressors vs catecholamines alone with atrial fibrillation in patients with distributive shock: A systematic review and meta-analysis. JAMA 2018; 319:1889–1900

233. Papoff P, Caresta E, Versacci P, et al: Beneficial effects of levo-simendan in infants with sepsis-associated cardiac dysfunction: Report of 2 cases. Pediatr Emerg Care 2012; 28:1062–1065

Page 44: Surviving Sepsis Campaign International Guidelines for the ... · In 2001, the Surviving Sepsis Campaign (SSC) was formed by the Society of Critical Care Medicine (SCCM), European

Copyright © 2020 by the Society of Critical Care Medicine and the World Federation of Pediatric Intensive and Critical Care Societies.Unauthorized reproduction of this article is prohibited

Online Special Article

Pediatric Critical Care Medicine www.pccmjournal.org e95

234. Rich N, West N, McMaster P, et al: Milrinone in meningococcal sepsis. Pediatr Crit Care Med 2003; 4:394–395

235. Pham T, Brochard LJ, Slutsky AS: Mechanical ventilation: State of the art. Mayo Clin Proc 2017; 92:1382–1400

236. Aubier M, Viires N, Syllie G, et al: Respiratory muscle contribution to lactic acidosis in low cardiac output. Am Rev Respir Dis 1982; 126:648–652

237. Cheifetz IM: Invasive and noninvasive pediatric mechanical ventila-tion. Respir Care 2003; 48:442–453; discussion 453–458

238. Ghuman AK, Newth CJ, Khemani RG: The association between the end tidal alveolar dead space fraction and mortality in pediatric acute hypoxemic respiratory failure. Pediatr Crit Care Med 2012; 13:11–15

239. Khemani RG, Smith L, Lopez-Fernandez YM, et al; Pediatric Acute Respiratory Distress syndrome Incidence and Epidemiology (PARDIE) Investigators; Pediatric Acute Lung Injury and Sepsis Investigators (PALISI) Network: Paediatric acute respiratory distress syndrome incidence and epidemiology (PARDIE): An international, observational study. Lancet Respir Med 2019; 7:115–128

240. Volpicelli G, Elbarbary M, Blaivas M, et al; International Liaison Committee on Lung Ultrasound (ILC-LUS) for International Consensus Conference on Lung Ultrasound (ICC-LUS): International evidence-based recommendations for point-of-care lung ultrasound. Intensive Care Med 2012; 38:577–591

241. Riviello ED, Kiviri W, Twagirumugabe T, et al: Hospital incidence and outcomes of the acute respiratory distress syndrome using the Kigali modification of the berlin definition. Am J Respir Crit Care Med 2016; 193:52–59

242. De Luca D, van Kaam AH, Tingay DG, et al: The montreux defi-nition of neonatal ARDS: Biological and clinical background be-hind the description of a new entity. Lancet Respir Med 2017; 5:657–666

243. See KC, Ong V, Tan YL, et al: Chest radiography versus lung ul-trasound for identification of acute respiratory distress syndrome: A retrospective observational study. Crit Care 2018; 22:203

244. Watt I, Ledingham IM: Mortality amongst multiple trauma patients admitted to an intensive therapy unit. Anaesthesia 1984; 39:973–981

245. den Brinker M, Joosten KF, Liem O, et al: Adrenal insufficiency in meningococcal sepsis: Bioavailable cortisol levels and impact of in-terleukin-6 levels and intubation with etomidate on adrenal function and mortality. J Clin Endocrinol Metab 2005; 90:5110–5117

246. Absalom A, Pledger D, Kong A: Adrenocortical function in critically ill patients 24 h after a single dose of etomidate. Anaesthesia 1999; 54:861–867

247. Schenarts CL, Burton JH, Riker RR: Adrenocortical dysfunction fol-lowing etomidate induction in emergency department patients. Acad Emerg Med 2001; 8:1–7

248. Hildreth AN, Mejia VA, Maxwell RA, et al: Adrenal suppression fol-lowing a single dose of etomidate for rapid sequence induction: A prospective randomized study. J Trauma 2008; 65:573–579

249. Jabre P, Combes X, Lapostolle F, et al; KETASED Collaborative Study Group: Etomidate versus ketamine for rapid sequence intuba-tion in acutely ill patients: A multicentre randomised controlled trial. Lancet 2009; 374:293–300

250. Muñoz-Bonet JI, Flor-Macián EM, Brines J, et al: Predictive factors for the outcome of noninvasive ventilation in pediatric acute respiratory failure. Pediatr Crit Care Med 2010; 11:675–680

251. James CS, Hallewell CP, James DP, et al: Predicting the success of non-invasive ventilation in preventing intubation and re-intubation in the paediatric intensive care unit. Intensive Care Med 2011; 37:1994–2001

252. Wolfler A, Calderini E, Iannella E, et al; Network of Pediatric Intensive Care Unit Study Group: Evolution of noninvasive mechanical ventila-tion use: A cohort study among Italian PICUs. Pediatr Crit Care Med 2015; 16:418–427

253. Abadesso C, Nunes P, Silvestre C, et al: Non-invasive ventilation in acute respiratory failure in children. Pediatr Rep 2012; 4:e16

254. Dohna-Schwake C, Stehling F, Tschiedel E, et al: Non-invasive venti-lation on a pediatric intensive care unit: Feasibility, efficacy, and pre-dictors of success. Pediatr Pulmonol 2011; 46:1114–1120

255. Piastra M, De Luca D, Marzano L, et al: The number of failing organs predicts non-invasive ventilation failure in children with ALI/ARDS. Intensive Care Med 2011; 37:1510–1516

256. Yaman A, Kendirli T, Ödek Ç, et al: Efficacy of noninvasive mechan-ical ventilation in prevention of intubation and reintubation in the pe-diatric intensive care unit. J Crit Care 2016; 32:175–181

257. Pancera CF, Hayashi M, Fregnani JH, et al: Noninvasive ventilation in immunocompromised pediatric patients: Eight years of experience in a pediatric oncology intensive care unit. J Pediatr Hematol Oncol 2008; 30:533–538

258. Peters MJ, Agbeko R, Davis P, et al; SCARF Study Investigators and the Pediatric Intensive Care Society Study Group (PICS-SG): Randomized study of early continuous positive airways pres-sure in acute respiratory failure in children with impaired immu-nity (SCARF) ISRCTN82853500. Pediatr Crit Care Med 2018; 19:939–948

259. Piastra M, De Luca D, Pietrini D, et al: Noninvasive pressure-support ventilation in immunocompromised children with ARDS: A feasibility study. Intensive Care Med 2009; 35:1420–1427

260. Essouri S, Carroll C; Pediatric Acute Lung Injury Consensus Conference Group: Noninvasive support and ventilation for pediatric acute respiratory distress syndrome: Proceedings from the Pediatric Acute Lung Injury Consensus Conference. Pediatr Crit Care Med 2015; 16:S102–S110

261. Kneyber MCJ, de Luca D, Calderini E, et al; section Respiratory Failure of the European Society for Paediatric and Neonatal Intensive Care: Recommendations for mechanical ventilation of critically ill children from the Paediatric Mechanical Ventilation Consensus Conference (PEMVECC). Intensive Care Med 2017; 43:1764–1780

262. Brower RG, Matthay MA, Morris A, et al; Acute Respiratory Distress Syndrome Network: Ventilation with lower tidal volumes as com-pared with traditional tidal volumes for acute lung injury and the acute respiratory distress syndrome. N Engl J Med 2000; 342: 1301–1308

263. Curley MA, Hibberd PL, Fineman LD, et al: Effect of prone position-ing on clinical outcomes in children with acute lung injury: A random-ized controlled trial. JAMA 2005; 294:229–237

264. Newth CJL, Sward KA, Khemani RG, et al; Eunice Kennedy Shriver National Institute for Child Health and Human Development Collaborative Pediatric Critical Care Research Network (CPCCRN): Variability in usual care mechanical ventilation for pediatric acute res-piratory distress syndrome: Time for a decision support protocol? Pediatr Crit Care Med 2017; 18:e521–e529

265. Erickson S, Schibler A, Numa A, et al; Paediatric Study Group; Australian and New Zealand Intensive Care Society: Acute lung injury in pediatric intensive care in Australia and New Zealand: A prospective, multicenter, observational study. Pediatr Crit Care Med 2007; 8:317–323

266. Flori HR, Glidden DV, Rutherford GW, et al: Pediatric acute lung in-jury: Prospective evaluation of risk factors associated with mortality. Am J Respir Crit Care Med 2005; 171:995–1001

267. Parvathaneni K, Belani S, Leung D, et al: Evaluating the performance of the pediatric acute lung injury consensus conference definition of acute respiratory distress syndrome. Pediatr Crit Care Med 2017; 18:17–25

268. Miller MP, Sagy M: Pressure characteristics of mechanical ventila-tion and incidence of pneumothorax before and after the implemen-tation of protective lung strategies in the management of pediatric patients with severe ARDS. Chest 2008; 134:969–973

269. Santschi M, Jouvet P, Leclerc F, et al; PALIVE Investigators; Pediatric Acute Lung Injury and Sepsis Investigators Network (PALISI); European Society of Pediatric and Neonatal Intensive Care (ESPNIC): Acute lung injury in children: Therapeutic practice and feasibility of international clinical trials. Pediatr Crit Care Med 2010; 11:681–689

270. Khemani RG, Markovitz BP, Curley MAQ: Characteristics of children intubated and mechanically ventilated in 16 PICUs. Chest 2009; 136:765–771

Page 45: Surviving Sepsis Campaign International Guidelines for the ... · In 2001, the Surviving Sepsis Campaign (SSC) was formed by the Society of Critical Care Medicine (SCCM), European

Copyright © 2020 by the Society of Critical Care Medicine and the World Federation of Pediatric Intensive and Critical Care Societies.Unauthorized reproduction of this article is prohibited

Copyright © 2020 by the Society of Critical Care Medicine and the World Federation of Pediatric Intensive and Critical Care Societies.Unauthorized reproduction of this article is prohibited

Weiss et al

e96 www.pccmjournal.org February 2020 • Volume 21 • Number 2

271. López-Fernández Y, Azagra AM, de la Oliva P, et al; Pediatric Acute Lung Injury Epidemiology and Natural History (PED-ALIEN) Network: Pediatric Acute Lung Injury Epidemiology and Natural History study: Incidence and outcome of the acute respiratory distress syndrome in children. Crit Care Med 2012; 40:3238–3245

272. Ingaramo OA, Ngo T, Khemani RG, et al: Impact of positive end-expiratory pressure on cardiac index measured by ultrasound car-diac output monitor*. Pediatr Crit Care Med 2014; 15:15–20

273. Guo F, Hao L, Zhen Q, et al: Multicenter study on the prognosis associated with respiratory support for children with acute hypoxic respiratory failure. Exp Ther Med 2016; 12:3227–3232

274. Yehya N, Thomas NJ: Disassociating lung mechanics and oxygena-tion in pediatric acute respiratory distress syndrome. Crit Care Med 2017; 45:1232–1239

275. Yehya N, Thomas NJ, Khemani RG: Risk stratification using oxygen-ation in the first 24 hours of pediatric acute respiratory distress syn-drome. Crit Care Med 2018; 46:619–624

276. Khemani RG, Parvathaneni K, Yehya N, et al: Positive end-expiratory pressure lower than the ARDS network protocol is associated with higher pediatric acute respiratory distress syndrome mortality. Am J Respir Crit Care Med 2018; 198:77–89

277. Halbertsma FJ, Vaneker M, Pickkers P, et al: A single recruitment maneuver in ventilated critically ill children can translocate pulmonary cytokines into the circulation. J Crit Care 2010; 25:10–15

278. Wolf GK, Gómez-Laberge C, Kheir JN, et al: Reversal of de-pendent lung collapse predicts response to lung recruitment in children with early acute lung injury. Pediatr Crit Care Med 2012; 13:509–515

279. Goligher EC, Hodgson CL, Adhikari NKJ, et al: Lung recruitment maneuvers for adult patients with acute respiratory distress syn-drome. A systematic review and meta-analysis. Ann Am Thorac Soc 2017; 14:S304–S311

280. Boriosi JP, Sapru A, Hanson JH, et al: Efficacy and safety of lung recruitment in pediatric patients with acute lung injury. Pediatr Crit Care Med 2011; 12:431–436

281. Duff JP, Rosychuk RJ, Joffe AR: The safety and efficacy of sustained inflations as a lung recruitment maneuver in pediatric intensive care unit patients. Intensive Care Med 2007; 33:1778–1786

282. Cavalcanti AB, Suzumura EA, Laranjeira LN, et al; Writing Group for the Alveolar Recruitment for Acute Respiratory Distress Syndrome Trial (ART) Investigators: Effect of lung recruitment and titrated positive end-expiratory pressure (PEEP) vs low PEEP on mortality in patients with acute respiratory distress syndrome: A randomized clinical trial. JAMA 2017; 318:1335–1345

283. Kang H, Yang H, Tong Z: Recruitment manoeuvres for adults with acute respiratory distress syndrome receiving mechanical ventilation: A systematic review and meta-analysis. J Crit Care 2019; 50:1–10

284. Santini A, Protti A, Langer T, et al: Prone position ameliorates lung elastance and increases functional residual capacity independently from lung recruitment. Intensive Care Med Exp 2015; 3:55

285. Sud S, Friedrich JO, Adhikari NK, et al: Effect of prone positioning during mechanical ventilation on mortality among patients with acute respiratory distress syndrome: A systematic review and meta-analy-sis. CMAJ 2014; 186:E381–E390

286. Sud S, Friedrich JO, Taccone P, et al: Prone ventilation reduces mor-tality in patients with acute respiratory failure and severe hypoxemia: Systematic review and meta-analysis. Intensive Care Med 2010; 36:585–599

287. Kornecki A, Frndova H, Coates AL, et al: 4A randomized trial of pro-longed prone positioning in children with acute respiratory failure. Chest 2001; 119:211–218

288. Macrae DJ, Field D, Mercier JC, et al: Inhaled nitric oxide therapy in neonates and children: Reaching a European consensus. Intensive Care Med 2004; 30:372–380

289. Day RW, Lynch JM, White KS, et al: Acute response to inhaled nitric oxide in newborns with respiratory failure and pulmonary hyperten-sion. Pediatrics 1996; 98:698–705

290. Dobyns EL, Cornfield DN, Anas NG, et al: Multicenter randomized controlled trial of the effects of inhaled nitric oxide therapy on gas

exchange in children with acute hypoxemic respiratory failure. J Pediatr 1999; 134:406–412

291. Bronicki RA, Fortenberry J, Schreiber M, et al: Multicenter random-ized controlled trial of inhaled nitric oxide for pediatric acute respira-tory distress syndrome. J Pediatr 2015; 166:365–369.e1

292. Karam O, Gebistorf F, Wetterslev J, et al: The effect of inhaled ni-tric oxide in acute respiratory distress syndrome in children and adults: A cochrane systematic review with trial sequential analysis. Anaesthesia 2017; 72:106–117

293. Fioretto JR, de Moraes MA, Bonatto RC, et al: Acute and sustained effects of early administration of inhaled nitric oxide to children with acute respiratory distress syndrome. Pediatr Crit Care Med 2004; 5:469–474

294. Tadphale SD, Rettiganti M, Gossett JM, et al: Is administration of nitric oxide during extracorporeal membrane oxygenation associ-ated with improved patient survival? Pediatr Crit Care Med 2016; 17:1080–1087

295. Dowell JC, Thomas NJ, Yehya N: Association of response to inhaled nitric oxide and duration of mechanical ventilation in pediatric acute respiratory distress syndrome. Pediatr Crit Care Med 2017; 18:1019–1026

296. Bhalla AK, Yehya N, Mack WJ, et al: The association between inhaled nitric oxide treatment and ICU mortality and 28-day ventilator-free days in pediatric acute respiratory distress syndrome. Crit Care Med 2018; 46:1803–1810

297. Gebistorf F, Karam O, Wetterslev J, et al: Inhaled nitric oxide for acute respiratory distress syndrome (ARDS) in children and adults. Cochrane Database Syst Rev 2016; 66:CD002787

298. Ferguson ND, Cook DJ, Guyatt GH, et al; OSCILLATE Trial Investigators; Canadian Critical Care Trials Group: High-frequency oscillation in early acute respiratory distress syndrome. N Engl J Med 2013; 368:795–805

299. Young D, Lamb SE, Shah S, et al; OSCAR Study Group: High-frequency oscillation for acute respiratory distress syndrome. N Engl J Med 2013; 368:806–813

300. Guo YX, Wang ZN, Li YT, et al: High-frequency oscillatory ventila-tion is an effective treatment for severe pediatric acute respiratory distress syndrome with refractory hypoxemia. Ther Clin Risk Manag 2016; 12:1563–1571

301. Bateman ST, Borasino S, Asaro LA, et al; RESTORE Study Investigators: Early high-frequency oscillatory ventilation in pediatric acute respiratory failure. A propensity score analysis. Am J Respir Crit Care Med 2016; 193:495–503

302. Arnold JH, Hanson JH, Toro-Figuero LO, et al: Prospective, random-ized comparison of high-frequency oscillatory ventilation and con-ventional mechanical ventilation in pediatric respiratory failure. Crit Care Med 1994; 22:1530–1539

303. Samransamruajkit R, Prapphal N, Deelodegenavong J, et al: Plasma soluble intercellular adhesion molecule-1 (sICAM-1) in pediatric ARDS during high frequency oscillatory ventilation: A predictor of mortality. Asian Pac J Allergy Immunol 2005; 23:181–188

304. Samransamruajkit R, Rassameehirun C, Pongsanon K, et al: A com-parison of clinical efficacy between high frequency oscillatory ven-tilation and conventional ventilation with lung volume recruitment in pediatric acute respiratory distress syndrome: A randomized con-trolled trial. Indian J Crit Care Med 2016; 20:72–77

305. Gainnier M, Roch A, Forel JM, et al: Effect of neuromuscular blocking agents on gas exchange in patients presenting with acute respiratory distress syndrome. Crit Care Med 2004; 32:113–119

306. Forel JM, Roch A, Marin V, et al: Neuromuscular blocking agents decrease inflammatory response in patients presenting with acute respiratory distress syndrome. Crit Care Med 2006; 34:2749–2757

307. Papazian L, Forel JM, Gacouin A, et al; ACURASYS Study Investigators: Neuromuscular blockers in early acute respiratory dis-tress syndrome. N Engl J Med 2010; 363:1107–1116

308. Wilsterman MEF, de Jager P, Blokpoel R, et al: Short-term effects of neuromuscular blockade on global and regional lung mechanics, oxygenation and ventilation in pediatric acute hypoxemic respiratory failure. Ann Intensive Care 2016; 6:103

Page 46: Surviving Sepsis Campaign International Guidelines for the ... · In 2001, the Surviving Sepsis Campaign (SSC) was formed by the Society of Critical Care Medicine (SCCM), European

Copyright © 2020 by the Society of Critical Care Medicine and the World Federation of Pediatric Intensive and Critical Care Societies.Unauthorized reproduction of this article is prohibited

Online Special Article

Pediatric Critical Care Medicine www.pccmjournal.org e97

309. Da Silva PS, Neto HM, de Aguiar VE, et al: Impact of sustained neuromuscular blockade on outcome of mechanically ventilated chil-dren. Pediatr Int 2010; 52:438–443

310. Wehling M: Specific, nongenomic actions of steroid hormones. Annu Rev Physiol 1997; 59:365–393

311. Seri I, Evans J: Controversies in the diagnosis and management of hypotension in the newborn infant. Curr Opin Pediatr 2001; 13:116–123

312. Munck A, Mendel DB, Smith LI, et al: Glucocorticoid receptors and actions. Am Rev Respir Dis 1990; 141:S2–10

313. Sasidharan P: Role of corticosteroids in neonatal blood pressure ho-meostasis. Clin Perinatol 1998; 25:723–740, xi

314. Yung M, Wilkins B, Norton L, et al; Paediatric Study Group; Australian and New Zealand Intensive Care Society: Glucose con-trol, organ failure, and mortality in pediatric intensive care. Pediatr Crit Care Med 2008; 9:147–152

315. Kwon S, Hermayer KL, Hermayer K: Glucocorticoid-induced hyper-glycemia. Am J Med Sci 2013; 345:274–277

316. Milan G, Romanello V, Pescatore F, et al: Regulation of autophagy and the ubiquitin-proteasome system by the FoxO transcriptional network during muscle atrophy. Nat Commun 2015; 6:6670

317. Steiner SJ, Noe JD, Denne SC: Corticosteroids increase protein breakdown and loss in newly diagnosed pediatric crohn disease. Pediatr Res 2011; 70:484–488

318. Costello JM, Graham DA, Morrow DF, et al: Risk factors for central line-associated bloodstream infection in a pediatric cardiac intensive care unit. Pediatr Crit Care Med 2009; 10:453–459

319. Waljee AK, Rogers MA, Lin P, et al: Short term use of oral corti-costeroids and related harms among adults in the United States: Population based cohort study. BMJ 2017; 357:j1415

320. El-Nawawy A, Khater D, Omar H, et al: Evaluation of early cortico-steroid therapy in management of pediatric septic shock in pediatric intensive care patients: A randomized clinical study. Pediatr Infect Dis J 2017; 36:155–159

321. Rygård SL, Butler E, Granholm A, et al: Low-dose corticosteroids for adult patients with septic shock: A systematic review with meta-analysis and trial sequential analysis. Intensive Care Med 2018; 44:1003–1016

322. Annane D, Sébille V, Charpentier C, et al: Effect of treatment with low doses of hydrocortisone and fludrocortisone on mortality in patients with septic shock. JAMA 2002; 288:862–871

323. Sprung CL, Annane D, Keh D, et al; CORTICUS Study Group: Hydrocortisone therapy for patients with septic shock. N Engl J Med 2008; 358:111–124

324. Annane D, Renault A, Brun-Buisson C, et al; CRICS-TRIGGERSEP Network: Hydrocortisone plus fludrocortisone for adults with septic shock. N Engl J Med 2018; 378:809–818

325. Venkatesh B, Finfer S, Cohen J, et al; ADRENAL Trial Investigators and the Australian–New Zealand Intensive Care Society Clinical Trials Group: Adjunctive glucocorticoid therapy in patients with septic shock. N Engl J Med 2018; 378:797–808

326. Yehya N, Vogiatzi MG, Thomas NJ, et al: Cortisol correlates with se-verity of illness and poorly reflects adrenal function in pediatric acute respiratory distress syndrome. J Pediatr 2016; 177:212–218.e1

327. Rochwerg B, Oczkowski SJ, Siemieniuk RAC, et al: Corticosteroids in sepsis: An updated systematic review and meta-analysis. Crit Care Med 2018; 46:1411–1420

328. Lamontagne F, Rochwerg B, Lytvyn L, et al: Corticosteroid therapy for sepsis: A clinical practice guideline. BMJ 2018; 362:k3284

329. Slusher T, Gbadero D, Howard C, et al: Randomized, placebo-controlled, double blinded trial of dexamethasone in African children with sepsis. Pediatr Infect Dis J 1996; 15:579–583

330. Valoor HT, Singhi S, Jayashree M: Low-dose hydrocortisone in pe-diatric septic shock: An exploratory study in a third world setting. Pediatr Crit Care Med 2009; 10:121–125

331. Menon K, McNally D, O’Hearn K, et al; Canadian Critical Care Trials Group: A randomized controlled trial of corticosteroids in pediatric septic shock: A pilot feasibility study. Pediatr Crit Care Med 2017; 18:505–512

332. Markovitz BP, Goodman DM, Watson RS, et al: A retrospective co-hort study of prognostic factors associated with outcome in pediatric severe sepsis: What is the role of steroids? Pediatr Crit Care Med 2005; 6:270–274

333. Zimmerman JJ, Williams MD: Adjunctive corticosteroid therapy in pediatric severe sepsis: Observations from the RESOLVE study. Pediatr Crit Care Med 2011; 12:2–8

334. Atkinson SJ, Cvijanovich NZ, Thomas NJ, et al: Corticosteroids and pediatric septic shock outcomes: A risk stratified analysis. PLoS One 2014; 9:e112702

335. Wong HR, Cvijanovich NZ, Anas N, et al: Developing a clinically fea-sible personalized medicine approach to pediatric septic shock. Am J Respir Crit Care Med 2015; 191:309–315

336. Menon K, McNally JD, Choong K, et al; Canadian Critical Care Trials Group STRIPES Investigators: A cohort study of pediatric shock: Frequency of corticosteriod use and association with clinical out-comes. Shock 2015; 44:402–409

337. Bornstein SR, Allolio B, Arlt W, et al: Diagnosis and treatment of primary adrenal insufficiency: An endocrine society clinical practice guideline. J Clin Endocrinol Metab 2016; 101:364–389

338. Broersen LH, Pereira AM, Jørgensen JO, et al: Adrenal insufficiency in corticosteroids use: Systematic review and meta-analysis. J Clin Endocrinol Metab 2015; 100:2171–2180

339. Agus MS, Steil GM, Wypij D, et al; SPECS Study Investigators: Tight glycemic control versus standard care after pediatric cardiac surgery. N Engl J Med 2012; 367:1208–1219

340. Agus MSD, Wypij D, Nadkarni, VM: Tight glycemic control in criti-cally ill children. N Engl J Med 2017; 376:e48

341. Macrae D, Grieve R, Allen E, et al; CHiP Investigators: A randomized trial of hyperglycemic control in pediatric intensive care. N Engl J Med 2014; 370:107–118

342. Vlasselaers D, Milants I, Desmet L, et al: Intensive insulin therapy for patients in paediatric intensive care: A prospective, randomised controlled study. Lancet 2009; 373:547–556

343. Jeschke MG, Kulp GA, Kraft R, et al: Intensive insulin therapy in se-verely burned pediatric patients: A prospective randomized trial. Am J Respir Crit Care Med 2010; 182:351–359

344. Chen L, Li T, Fang F, et al: Tight glycemic control in critically ill pe-diatric patients: A systematic review and meta-analysis. Crit Care 2018; 22:57

345. Zhao Y, Wu Y, Xiang B: Tight glycemic control in critically ill pedi-atric patients: A meta-analysis and systematic review of randomized controlled trials. Pediatr Res 2018; 84:22–27

346. McKinlay CJD, Alsweiler JM, Anstice NS, et al; Children With Hypoglycemia and Their Later Development (CHYLD) Study Team: Association of neonatal glycemia with neurodevelopmental out-comes at 4.5 years. JAMA Pediatr 2017; 171:972–983

347. Mesotten D, Gielen M, Sterken C, et al: Neurocognitive develop-ment of children 4 years after critical illness and treatment with tight glucose control: A randomized controlled trial. JAMA 2012; 308:1641–1650

348. Sadhwani A, Asaro LA, Goldberg C, et al: Impact of tight glycemic control on neurodevelopmental outcomes at 1 year of age for chil-dren with congenital heart disease: A randomized controlled trial. J Pediatr 2016; 174:193–198.e2

349. Faustino EVS, Hirshberg EL, Asaro LA, et al; Heart And Lung Failure-Pediatric INsulin Titration (HALF-PINT) Study Investigators: Short-term adverse outcomes associated with hypoglycemia in critically ill children. Crit Care Med 2019; 47:706–714

350. Forsythe RM, Wessel CB, Billiar TR, et al: Parenteral calcium for intensive care unit patients. Cochrane Database Syst Rev 2008; (4):CD006163

351. Dotson B, Larabell P, Patel JU, et al: Calcium administration is asso-ciated with adverse outcomes in critically ill patients receiving paren-teral nutrition: Results from a natural experiment created by a calcium gluconate shortage. Pharmacotherapy 2016; 36:1185–1190

352. Dias CR, Leite HP, Nogueira PC, et al: Ionized hypocalcemia is an early event and is associated with organ dysfunction in children admitted to the intensive care unit. J Crit Care 2013; 28:810–815

Page 47: Surviving Sepsis Campaign International Guidelines for the ... · In 2001, the Surviving Sepsis Campaign (SSC) was formed by the Society of Critical Care Medicine (SCCM), European

Copyright © 2020 by the Society of Critical Care Medicine and the World Federation of Pediatric Intensive and Critical Care Societies.Unauthorized reproduction of this article is prohibited

Copyright © 2020 by the Society of Critical Care Medicine and the World Federation of Pediatric Intensive and Critical Care Societies.Unauthorized reproduction of this article is prohibited

Weiss et al

e98 www.pccmjournal.org February 2020 • Volume 21 • Number 2

353. Hebbar K, Rigby MR, Felner EI, et al: Neuroendocrine dysfunction in pediatric critical illness. Pediatr Crit Care Med 2009; 10:35–40

354. Shih JL, Agus MS: Thyroid function in the critically ill newborn and child. Curr Opin Pediatr 2009; 21:536–540

355. Marwali EM, Boom CE, Sakidjan I, et al: Oral triiodothyronine normal-izes triiodothyronine levels after surgery for pediatric congenital heart disease*. Pediatr Crit Care Med 2013; 14:701–708

356. Marwali EM, Boom CE, Budiwardhana N, et al: Oral triiodothyronine for infants and children undergoing cardiopulmonary bypass. Ann Thorac Surg 2017; 104:688–695

357. Smith LM, Leake RD, Berman N, et al: Postnatal thyroxine supple-mentation in infants less than 32 weeks’ gestation: Effects on pul-monary morbidity. J Perinatol 2000; 20:427–431

358. Lee BH, Inui D, Suh GY, et al; Fever and Antipyretic in Critically ill patients Evaluation (FACE) Study Group: Association of body temperature and antipyretic treatments with mortality of critically ill patients with and without sepsis: Multi-centered prospective obser-vational study. Crit Care 2012; 16:R33

359. Walter EJ, Hanna-Jumma S, Carraretto M, et al: The pathophysiolog-ical basis and consequences of fever. Crit Care 2016; 20:200

360. Sullivan JE, Farrar HC; Section on Clinical Pharmacology and Therapeutics, Committee on Drugs: Fever and antipyretic use in chil-dren. Pediatrics 2011; 127:580–587

361. Drewry AM, Ablordeppey EA, Murray ET, et al: Antipyretic therapy in critically ill septic patients: A systematic review and meta-analysis. Crit Care Med 2017; 45:806–813

362. Prakash V, Parameswaran N, Biswal N: Early versus late enteral feed-ing in critically ill children: A randomized controlled trial. Intensive Care Med 2016; 42:481–482

363. Rice TW, Mogan S, Hays MA, et al: Randomized trial of initial trophic versus full-energy enteral nutrition in mechanically ventilated patients with acute respiratory failure. Crit Care Med 2011; 39: 967–974

364. Meyer R, Harrison S, Sargent S, et al: The impact of enteral feeding protocols on nutritional support in critically ill children. J Hum Nutr Diet 2009; 22:428–436

365. Petrillo-Albarano T, Pettignano R, Asfaw M, et al: Use of a feeding protocol to improve nutritional support through early, aggressive, en-teral nutrition in the pediatric intensive care unit. Pediatr Crit Care Med 2006; 7:340–344

366. Yoshimura S, Miyazu M, Yoshizawa S, et al: Efficacy of an enteral feeding protocol for providing nutritional support after paediatric car-diac surgery. Anaesth Intensive Care 2015; 43:587–593

367. Hamilton S, McAleer DM, Ariagno K, et al: A stepwise enteral nutri-tion algorithm for critically ill children helps achieve nutrient delivery goals*. Pediatr Crit Care Med 2014; 15:583–589

368. Panchal AK, Manzi J, Connolly S, et al: Safety of enteral feedings in critically ill children receiving vasoactive agents. JPEN J Parenter Enteral Nutr 2016; 40:236–241

369. King W, Petrillo T, Pettignano R: Enteral nutrition and cardiovascular medications in the pediatric intensive care unit. JPEN J Parenter Enteral Nutr 2004; 28:334–338

370. López-Herce J, Santiago MJ, Sánchez C, et al: Risk factors for gas-trointestinal complications in critically ill children with transpyloric en-teral nutrition. Eur J Clin Nutr 2008; 62:395–400

371. Arabi YM, Aldawood AS, Haddad SH, et al; PermiT Trial Group: Permissive underfeeding or standard enteral feeding in critically ill adults. N Engl J Med 2015; 372:2398–2408

372. Fivez T, Kerklaan D, Mesotten D, et al: Early versus late parenteral nu-trition in critically ill children. N Engl J Med 2016; 374:1111–1122

373. van Puffelen E, Vanhorebeek I, Joosten KFM, et al: Early versus late parenteral nutrition in critically ill, term neonates: A preplanned sec-ondary subgroup analysis of the PEPaNIC multicentre, randomised controlled trial. Lancet Child Adolesc Health 2018; 2:505–515

374. van Puffelen E, Hulst JM, Vanhorebeek I, et al: Outcomes of delaying parenteral nutrition for 1 week vs initiation within 24 hours among undernourished children in pediatric intensive care: A subanalysis of the PEPaNIC randomized clinical trial. JAMA Netw Open 2018; 1:e182668

375. Verstraete S, Verbruggen SC, Hordijk JA, et al: Long-term develop-mental effects of withholding parenteral nutrition for 1 week in the paediatric intensive care unit: A 2-year follow-up of the PEPaNIC international, randomised, controlled trial. Lancet Respir Med 2019; 7:141–153

376. Doig GS, Simpson F, Sweetman EA, et al; Early PN Investigators of the ANZICS Clinical Trials Group: Early parenteral nutrition in critically ill patients with short-term relative contraindications to early enteral nutrition: A randomized controlled trial. JAMA 2013; 309:2130–2138

377. Casaer MP, Mesotten D, Hermans G, et al: Early versus late paren-teral nutrition in critically ill adults. N Engl J Med 2011; 365:506–517

378. Singer P, Anbar R, Cohen J, et al: The tight calorie control study (TICACOS): A prospective, randomized, controlled pilot study of nu-tritional support in critically ill patients. Intensive Care Med 2011; 37:601–609

379. Briassoulis G, Filippou O, Hatzi E, et al: Early enteral administration of immunonutrition in critically ill children: Results of a blinded ran-domized controlled clinical trial. Nutrition 2005; 21:799–807

380. Carcillo JA, Dean JM, Holubkov R, et al; Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD) Collaborative Pediatric Critical Care Research Network (CPCCRN): The randomized comparative pediatric critical illness stress-induced immune suppression (CRISIS) prevention trial. Pediatr Crit Care Med 2012; 13:165–173

381. Al-Biltagi MA, Abo-Elezz AA, Abd-Elhafez MA, et al: Beneficial effects of omega-3 supplement to the enteral feeding in children with mild to moderate sepsis. J Intensive Care Med 2017; 32:212–217

382. Tume LN, Bickerdike A, Latten L, et al: Routine gastric residual volume measurement and energy target achievement in the PICU: A comparison study. Eur J Pediatr 2017; 176:1637–1644

383. Meert KL, Daphtary KM, Metheny NA: Gastric vs small-bowel feed-ing in critically ill children receiving mechanical ventilation: A random-ized controlled trial. Chest 2004; 126:872–878

384. Kamat P, Favaloro-Sabatier J, Rogers K, et al: Use of methylene blue spectrophotometry to detect subclinical aspiration in enter-ally fed intubated pediatric patients. Pediatr Crit Care Med 2008; 9:299–303

385. Sönmez Düzkaya D, Yildiz S: Effect of two different feeding methods on preventing ventilator associated pneumonia in the paediatric in-tensive care unit (PICU): A randomised controlled study. Aust Crit Care 2016; 29:139–145

386. Lewis K, Alqahtani Z, Mcintyre L, et al: The efficacy and safety of prokinetic agents in critically ill patients receiving enteral nutrition: A systematic review and meta-analysis of randomized trials. Crit Care 2016; 20:259

387. Yap YG, Camm AJ: Drug induced QT prolongation and torsades de pointes. Heart 2003; 89:1363–1372

388. Freeman BD, Dixon DJ, Coopersmith CM, et al: Pharmacoepidemiology of QT-interval prolonging drug administration in critically ill patients. Pharmacoepidemiol Drug Saf 2008; 17:971–981

389. Ray WA, Murray KT, Meredith S, et al: Oral erythromycin and the risk of sudden death from cardiac causes. N Engl J Med 2004; 351:1089–1096

390. Stadtman TC: Selenocysteine. Annu Rev Biochem 1996; 65:83–100

391. Iglesias SB, Leite HP, Paes AT, et al: Low plasma selenium concen-trations in critically ill children: The interaction effect between inflam-mation and selenium deficiency. Crit Care 2014; 18:R101

392. Loui A, Raab A, Braetter P, et al: Selenium status in term and pre-term infants during the first months of life. Eur J Clin Nutr 2008; 62:349–355

393. Sammalkorpi K, Valtonen V, Alfthan G, et al: Serum selenium in acute infections. Infection 1988; 16:222–224

394. Asci A, Surmeli-Onay O, Erkekoglu P, et al: Oxidant and antioxidant status in neonatal proven and clinical sepsis according to selenium status. Pediatr Int 2015; 57:1131–1137

395. Mertens K, Lowes DA, Webster NR, et al: Low zinc and selenium concentrations in sepsis are associated with oxidative damage and inflammation. Br J Anaesth 2015; 114:990–999

Page 48: Surviving Sepsis Campaign International Guidelines for the ... · In 2001, the Surviving Sepsis Campaign (SSC) was formed by the Society of Critical Care Medicine (SCCM), European

Copyright © 2020 by the Society of Critical Care Medicine and the World Federation of Pediatric Intensive and Critical Care Societies.Unauthorized reproduction of this article is prohibited

Online Special Article

Pediatric Critical Care Medicine www.pccmjournal.org e99

396. Darlow BA, Austin NC: Selenium supplementation to prevent short-term morbidity in preterm neonates. Cochrane Database Syst Rev 2003; (4):CD003312

397. Alhazzani W, Almasoud A, Jaeschke R, et al: Small bowel feeding and risk of pneumonia in adult critically ill patients: A systematic review and meta-analysis of randomized trials. Crit Care 2013; 17:R127

398. Barbosa E, Moreira EA, Goes JE, et al: Pilot study with a glutamine-supplemented enteral formula in critically ill infants. Rev Hosp Clin Fac Med Sao Paulo 1999; 54:21–24

399. Briassoulis G, Filippou O, Kanariou M, et al: Comparative effects of early randomized immune or non-immune-enhancing enteral nutrition on cytokine production in children with septic shock. Intensive Care Med 2005; 31:851–858

400. Heyland D, Muscedere J, Wischmeyer PE, et al; Canadian Critical Care Trials Group: A randomized trial of glutamine and antioxidants in critically ill patients. N Engl J Med 2013; 368:1489–1497

401. Jordan I, Balaguer M, Esteban ME, et al: Glutamine effects on heat shock protein 70 and interleukines 6 and 10: Randomized trial of glutamine supplementation versus standard parenteral nutrition in critically ill children. Clin Nutr 2016; 35:34–40

402. Novak F, Heyland DK, Avenell A, et al: Glutamine supplementation in serious illness: A systematic review of the evidence. Crit Care Med 2002; 30:2022–2029

403. van Zanten AR, Sztark F, Kaisers UX, et al: High-protein enteral nu-trition enriched with immune-modulating nutrients vs standard high-protein enteral nutrition and nosocomial infections in the ICU: A randomized clinical trial. JAMA 2014; 312:514–524

404. Wernerman J, Kirketeig T, Andersson B, et al; Scandinavian Critical Care Trials Group: Scandinavian glutamine trial: A pragmatic multi-centre randomised clinical trial of intensive care unit patients. Acta Anaesthesiol Scand 2011; 55:812–818

405. Beale RJ, Sherry T, Lei K, et al: Early enteral supplementation with key pharmaconutrients improves sequential organ failure assess-ment score in critically ill patients with sepsis: Outcome of a randomized, controlled, double-blind trial. Crit Care Med 2008; 36:131–144

406. Fuentes-Orozco C, Anaya-Prado R, González-Ojeda A, et al: L-alanyl-L-glutamine-supplemented parenteral nutrition improves infectious morbidity in secondary peritonitis. Clin Nutr 2004; 23:13–21

407. Koksal GM, Erbabacan E, Tunali Y, et al: The effects of intravenous, enteral and combined administration of glutamine on malnutrition in sepsis: A randomized clinical trial. Asia Pac J Clin Nutr 2014; 23:34–40

408. Bollhalder L, Pfeil AM, Tomonaga Y, et al: A systematic literature re-view and meta-analysis of randomized clinical trials of parenteral glu-tamine supplementation. Clin Nutr 2013; 32:213–223

409. Tao KM, Li XQ, Yang LQ, et al: Glutamine supplementation for criti-cally ill adults. Cochrane Database Syst Rev 2014; (9):CD010050

410. Tubman TR, Thompson SW, McGuire W: Glutamine supplementa-tion to prevent morbidity and mortality in preterm infants. Cochrane Database Syst Rev 2008; (1):CD001457

411. Carcillo JA, Dean JM, Holubkov R, et al; Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD) Collaborative Pediatric Critical Care Research Network (CPCCRN): Interaction between 2 nutraceutical treatments and host immune status in the pediatric critical illness stress-induced immune sup-pression comparative effectiveness trial. JPEN J Parenter Enteral Nutr 2017; 41:1325–1335

412. Barbul A, Uliyargoli A: Use of exogenous arginine in multiple organ dysfunction syndrome and sepsis. Crit Care Med 2007; 35:S564–S567

413. Bertolini G, Iapichino G, Radrizzani D, et al: Early enteral immunonu-trition in patients with severe sepsis: Results of an interim analysis of a randomized multicentre clinical trial. Intensive Care Med 2003; 29:834–840

414. Suchner U, Kuhn KS, Fürst P: The scientific basis of immunonutri-tion. Proc Nutr Soc 2000; 59:553–563

415. Argaman Z, Young VR, Noviski N, et al: Arginine and nitric oxide me-tabolism in critically ill septic pediatric patients. Crit Care Med 2003; 31:591–597

416. Bower RH, Cerra FB, Bershadsky B, et al: Early enteral administra-tion of a formula (impact) supplemented with arginine, nucleotides, and fish oil in intensive care unit patients: Results of a multicenter, prospective, randomized, clinical trial. Crit Care Med 1995; 23:436–449

417. Caparrós T, Lopez J, Grau T: Early enteral nutrition in critically ill patients with a high-protein diet enriched with arginine, fiber, and antioxidants compared with a standard high-protein diet. The effect on nosocomial infections and outcome. JPEN J Parenter Enteral Nutr 2001; 25:299–308; discussion 308–309

418. Galbán C, Montejo JC, Mesejo A, et al: An immune-enhancing enteral diet reduces mortality rate and episodes of bacteremia in septic intensive care unit patients. Crit Care Med 2000; 28:643–648

419. Santora R, Kozar RA: Molecular mechanisms of pharmaconutrients. J Surg Res 2010; 161:288–294

420. Kieft H, Roos AN, van Drunen JD, et al: Clinical outcome of immu-nonutrition in a heterogeneous intensive care population. Intensive Care Med 2005; 31:524–532

421. Taylor BE, McClave SA, Martindale RG, et al; Society of Critical Care Medicine; American Society of Parenteral and Enteral Nutrition: Guidelines for the provision and assessment of nutrition support therapy in the adult critically ill patient: Society of Critical Care Medicine (SCCM) and American Society for Parenteral and Enteral Nutrition (A.S.P.E.N.). Crit Care Med 2016; 44:390–438

422. L Langlois P, Lamontagne F: Vitamin C for the critically ill: Is the evi-dence strong enough? Nutrition 2019; 60:185–190

423. Marik PE: Vitamin C for the treatment of sepsis: The scientific ra-tionale. Pharmacol Ther 2018; 189:63–70

424. Teng J, Pourmand A, Mazer-Amirshahi M: Vitamin C: The next step in sepsis management? J Crit Care 2018; 43:230–234

425. Carr AC, Rosengrave PC, Bayer S, et al: Hypovitaminosis C and vitamin C deficiency in critically ill patients despite recommended enteral and parenteral intakes. Crit Care 2017; 21:300

426. Fowler AA 3rd, Syed AA, Knowlson S, et al; Medical Respiratory Intensive Care Unit Nursing: Phase I safety trial of intravenous ascorbic acid in patients with severe sepsis. J Transl Med 2014; 12:32

427. Zabet MH, Mohammadi M, Ramezani M, et al: Effect of high-dose ascorbic acid on vasopressor’s requirement in septic shock. J Res Pharm Pract 2016; 5:94–100

428. Moskowitz A, Andersen LW, Huang DT, et al: Ascorbic acid, cor-ticosteroids, and thiamine in sepsis: A review of the biologic ra-tionale and the present state of clinical evaluation. Crit Care 2018; 22:283

429. Marik PE, Khangoora V, Rivera R, et al: Hydrocortisone, vitamin C, and thiamine for the treatment of severe sepsis and septic shock: A Retrospective Before-After Study. Chest 2017; 151:1229–1238

430. Donnino MW, Andersen LW, Chase M, et al; Center for Resuscitation Science Research Group: Randomized, double-blind, placebo-con-trolled trial of thiamine as a metabolic resuscitator in septic shock: A pilot study. Crit Care Med 2016; 44:360–367

431. Lima LF, Leite HP, Taddei JA: Low blood thiamine concentrations in children upon admission to the intensive care unit: Risk factors and prognostic significance. Am J Clin Nutr 2011; 93:57–61

432. Costa NA, Gut AL, de Souza Dorna M, et al: Serum thiamine con-centration and oxidative stress as predictors of mortality in patients with septic shock. J Crit Care 2014; 29:249–252

433. McNally JD, Menon K, Chakraborty P, et al; Canadian Critical Care Trials Group: The association of vitamin D status with pediatric crit-ical illness. Pediatrics 2012; 130:429–436

434. Madden K, Feldman HA, Smith EM, et al: Vitamin D deficiency in critically ill children. Pediatrics 2012; 130:421–428

435. Sankar J, Lotha W, Ismail J, et al: Vitamin D deficiency and length of pediatric intensive care unit stay: A prospective observational study. Ann Intensive Care 2016; 6:3

Page 49: Surviving Sepsis Campaign International Guidelines for the ... · In 2001, the Surviving Sepsis Campaign (SSC) was formed by the Society of Critical Care Medicine (SCCM), European

Copyright © 2020 by the Society of Critical Care Medicine and the World Federation of Pediatric Intensive and Critical Care Societies.Unauthorized reproduction of this article is prohibited

Weiss et al

e100 www.pccmjournal.org February 2020 • Volume 21 • Number 2

436. Ponnarmeni S, Kumar Angurana S, Singhi S, et al: Vitamin D defi-ciency in critically ill children with sepsis. Paediatr Int Child Health 2016; 36:15–21

437. Onwuneme C, Carroll A, Doherty D, et al: Inadequate vitamin D lev-els are associated with culture positive sepsis and poor outcomes in paediatric intensive care. Acta Paediatr 2015; 104:e433–e438

438. Madden K, Feldman HA, Chun RF, et al: Critically ill children have low vitamin D-binding protein, influencing bioavailability of vitamin D. Ann Am Thorac Soc 2015; 12:1654–1661

439. McNally JD, Iliriani K, Pojsupap S, et al: Rapid normaliza-tion of vitamin D levels: A meta-analysis. Pediatrics 2015; 135:e152–e166

440. McNally D, Amrein K, O’Hearn K, et al; Canadian Critical Care Trials Group: Study protocol for a phase II dose evaluation random-ized controlled trial of cholecalciferol in critically ill children with vitamin D deficiency (VITdAL-PICU study). Pilot Feasibility Stud 2017; 3:70

441. McNally JD: Vitamin D deficiency in pediatric critical illness: Time to move on from observational studies? Rev Chil Pediatr 2016; 87:439–441

442. Marcinowska-Suchowierska E, Kupisz-Urbańska M, Łukaszkiewicz J, et al: Vitamin D toxicity-a clinical perspective. Front Endocrinol (Lausanne) 2018; 9:550

443. Narsaria P, Sankar J, Lodha R: Fatal outcome of accidental vitamin D overdose. Indian J Pediatr 2016; 83:1040

444. Misra M, Pacaud D, Petryk A, et al; Drug and Therapeutics Committee of the Lawson Wilkins Pediatric Endocrine Society: Vitamin D defi-ciency in children and its management: Review of current knowledge and recommendations. Pediatrics 2008; 122:398–417

445. Karam O, Tucci M, Ducruet T, et al; Canadian Critical Care Trials Group; PALISI Network: Red blood cell transfusion thresholds in pediatric patients with sepsis. Pediatr Crit Care Med 2011; 12:512–518

446. Lacroix J, Hébert PC, Hutchison JS, et al; TRIPICU Investigators; Canadian Critical Care Trials Group; Pediatric Acute Lung Injury and Sepsis Investigators Network: Transfusion strategies for patients in pediatric intensive care units. N Engl J Med 2007; 356:1609–1619

447. Hébert PC, Wells G, Blajchman MA, et al: A multicenter, randomized, controlled clinical trial of transfusion requirements in critical care. Transfusion requirements in Critical Care Investigators, Canadian Critical Care Trials Group. N Engl J Med 1999; 340:409–417

448. Muszynski JA, Guzzetta NA, Hall MW, et al; Pediatric Critical Care Transfusion and Anemia Expertise Initiative (TAXI); Pediatric Critical Care Blood Research Network (Blood Net), and the Pediatric Acute Lung Injury and Sepsis Investigators (PALISI) Network: Recommendations on RBC transfusions for critically ill children with nonhemorrhagic shock from the pediatric critical care transfu-sion and anemia expertise initiative. Pediatr Crit Care Med 2018; 19:S121–S126

449. Sankar J, Sankar MJ, Suresh CP, et al: Early goal-directed therapy in pediatric septic shock: Comparison of outcomes “with” and “without” intermittent superior venacaval oxygen saturation moni-toring: A prospective cohort study*. Pediatr Crit Care Med 2014; 15:e157–e167

450. Holst LB, Haase N, Wetterslev J, et al; TRISS Trial Group; Scandinavian Critical Care Trials Group: Lower versus higher hemo-globin threshold for transfusion in septic shock. N Engl J Med 2014; 371:1381–1391

451. Du Pont-Thibodeau G, Tucci M, Robitaille N, et al: Platelet trans-fusions in pediatric intensive care. Pediatr Crit Care Med 2016; 17:e420–e429

452. Curley A, Stanworth SJ, Willoughby K, et al; PlaNeT2 MATISSE Collaborators: Randomized trial of platelet-transfusion thresholds in neonates. N Engl J Med 2019; 380:242–251

453. Pieters BJ, Conley L, Weiford J, et al: Prophylactic versus reactive transfusion of thawed plasma in patients undergoing surgical repair of craniosynostosis: A randomized clinical trial. Paediatr Anaesth 2015; 25:279–287

454. Yang L, Stanworth S, Hopewell S, et al: Is fresh-frozen plasma clini-cally effective? An update of a systematic review of randomized con-trolled trials. Transfusion 2012; 52:1673–1686; quiz 1673

455. Church GD, Matthay MA, Liu K, et al: Blood product transfusions and clinical outcomes in pediatric patients with acute lung injury. Pediatr Crit Care Med 2009; 10:297–302

456. Karam O, Lacroix J, Robitaille N, et al: Association between plasma transfusions and clinical outcome in critically ill children: A prospec-tive observational study. Vox Sang 2013; 104:342–349

457. Abdel-Wahab OI, Healy B, Dzik WH: Effect of fresh-frozen plasma transfusion on prothrombin time and bleeding in patients with mild coagulation abnormalities. Transfusion 2006; 46:1279–1285

458. Rimmer E, Houston BL, Kumar A, et al: The efficacy and safety of plasma exchange in patients with sepsis and septic shock: A system-atic review and meta-analysis. Crit Care 2014; 18:699

459. Schwartz J, Padmanabhan A, Aqui N, et al: Guidelines on the use of therapeutic apheresis in clinical practice-evidence-based approach from the Writing Committee of the American Society for Apheresis: The seventh special issue. J Clin Apher 2016; 31:149–162

460. Nguyen TC, Han YY, Kiss JE, et al: Intensive plasma exchange increases a disintegrin and metalloprotease with thrombospondin motifs-13 activity and reverses organ dysfunction in children with thrombocytopenia-associated multiple organ failure. Crit Care Med 2008; 36:2878–2887

461. Carcillo JA, Berg RA, Wessel D, et al; Eunice Kennedy Shriver National Institute of Child Health and Human Development Collaborative Pediatric Critical Care Research Network.: A multi-center network assessment of three inflammation phenotypes in pe-diatric sepsis-induced multiple organ failure. Pediatr Crit Care Med 2019; 20:1137–1146

462. Nguyen TC, Liu A, Liu L, et al: Acquired ADAMTS-13 deficiency in pediatric patients with severe sepsis. Haematologica 2007; 92:121–124

463. Bernardo A, Ball C, Nolasco L, et al: Effects of inflammatory cyto-kines on the release and cleavage of the endothelial cell-derived ultralarge von Willebrand factor multimers under flow. Blood 2004; 104:100–106

464. Ono T, Mimuro J, Madoiwa S, et al: Severe secondary deficiency of von Willebrand factor-cleaving protease (ADAMTS13) in patients with sepsis-induced disseminated intravascular coagula-tion: Its correlation with development of renal failure. Blood 2006; 107:528–534

465. Crawley JT, Lam JK, Rance JB, et al: Proteolytic inactiva-tion of ADAMTS13 by thrombin and plasmin. Blood 2005; 105:1085–1093

466. Studt JD, Kremer Hovinga JA, Antoine G, et al: Fatal congenital thrombotic thrombocytopenic purpura with apparent ADAMTS13 inhibitor: In vitro inhibition of ADAMTS13 activity by hemoglobin. Blood 2005; 105:542–544

467. Nolasco LH, Turner NA, Bernardo A, et al: Hemolytic uremic syn-drome-associated Shiga toxins promote endothelial-cell secretion and impair ADAMTS13 cleavage of unusually large von Willebrand factor multimers. Blood 2005; 106:4199–4209

468. Tsai HM, Lian EC: Antibodies to von Willebrand factor-cleaving pro-tease in acute thrombotic thrombocytopenic purpura. N Engl J Med 1998; 339:1585–1594

469. Fortenberry JD, Nguyen T, Grunwell JR, et al; Thrombocytopenia-Associated Multiple Organ Failure (TAMOF) Network Study Group: Therapeutic plasma exchange in children with thrombocytopenia-associated multiple organ failure: The Thrombocytopenia-Associated Multiple Organ Failure Network prospective experience. Crit Care Med 2019; 47:e173–e181

470. Sevketoglu E, Yildizdas D, Horoz OO, et al: Use of therapeutic plasma exchange in children with thrombocytopenia-associated multiple organ failure in the Turkish thrombocytopenia-associ-ated multiple organ failure network. Pediatr Crit Care Med 2014; 15:e354–e359

471. Rock GA, Shumak KH, Buskard NA, et al: Comparison of plasma exchange with plasma infusion in the treatment of thrombotic

Page 50: Surviving Sepsis Campaign International Guidelines for the ... · In 2001, the Surviving Sepsis Campaign (SSC) was formed by the Society of Critical Care Medicine (SCCM), European

Copyright © 2020 by the Society of Critical Care Medicine and the World Federation of Pediatric Intensive and Critical Care Societies.Unauthorized reproduction of this article is prohibited

Online Special Article

Pediatric Critical Care Medicine www.pccmjournal.org e101

thrombocytopenic purpura. Canadian Apheresis Study Group. N Engl J Med 1991; 325:393–397

472. Guo XH, Sun YF, Han SZ, et al: Continuous blood purification in children with severe sepsis. J Biol Regul Homeost Agents 2017; 31:389–394

473. Sutherland SM, Zappitelli M, Alexander SR, et al: Fluid overload and mortality in children receiving continuous renal replacement therapy: The prospective pediatric continuous renal replacement therapy reg-istry. Am J Kidney Dis 2010; 55:316–325

474. Gulla KM, Sachdev A, Gupta D, et al: Continuous renal replacement therapy in children with severe sepsis and multiorgan dysfunction - a pilot study on timing of initiation. Indian J Crit Care Med 2015; 19:613–617

475. Borthwick EM, Hill CJ, Rabindranath KS, et al: High-volume haemo-filtration for sepsis in adults. Cochrane Database Syst Rev 2017; 1:CD008075

476. Miao H, Wang F, Xiong X, et al: Clinical benefits of high-volume hemofiltration in critically ill pediatric patients with severe sepsis: A Retrospective Cohort Study. Blood Purif 2018; 45:18–27

477. Nasr VG, Raman L, Barbaro RP, et al: Highlights from the extracor-poreal life support organization registry: 2006-2017. ASAIO J 2019; 65:537–544

478. Maclaren G, Butt W, Best D, et al: Extracorporeal membrane ox-ygenation for refractory septic shock in children: One institution’s experience. Pediatr Crit Care Med 2007; 8:447–451

479. Ruth A, McCracken CE, Fortenberry JD, et al: Extracorporeal thera-pies in pediatric severe sepsis: Findings from the pediatric health-care information system. Crit Care 2015; 19:397

480. Skinner SC, Iocono JA, Ballard HO, et al: Improved survival in venovenous vs venoarterial extracorporeal membrane oxygen-ation for pediatric noncardiac sepsis patients: A study of the Extracorporeal Life Support Organization registry. J Pediatr Surg 2012; 47:63–67

481. Barbaro RP, Xu Y, Borasino S, et al; RESTORE Study Investigators *: Does extracorporeal membrane oxygenation improve survival in pediatric acute respiratory failure? Am J Respir Crit Care Med 2018; 197:1177–1186

482. Curley MA, Wypij D, Watson RS, et al; RESTORE Study Investigators and the Pediatric Acute Lung Injury and Sepsis Investigators Network: Protocolized sedation vs usual care in pediatric patients mechanically ventilated for acute respiratory failure: A randomized clinical trial. JAMA 2015; 313:379–389

483. Bailly DK, Reeder RW, Winder M, et al; Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD) Collaborative Pediatric Critical Care Research Network (CPCCRN): Development of the pediatric extracorporeal membrane oxygenation prediction model for risk-adjusting mortality. Pediatr Crit Care Med 2019; 20:426–434

484. Jen HC, Shew SB: Hospital readmissions and survival after nonneo-natal pediatric ECMO. Pediatrics 2010; 125:1217–1223

485. Barbaro RP, Odetola FO, Kidwell KM, et al: Association of hospital-level volume of extracorporeal membrane oxygenation cases and mortality. Analysis of the extracorporeal life support organization reg-istry. Am J Respir Crit Care Med 2015; 191:894–901

486. MacLaren G, Butt W, Best D, et al: Central extracorporeal mem-brane oxygenation for refractory pediatric septic shock. Pediatr Crit Care Med 2011; 12:133–136

487. Bréchot N, Luyt CE, Schmidt M, et al: Venoarterial extracorporeal membrane oxygenation support for refractory cardiovascular dys-function during severe bacterial septic shock. Crit Care Med 2013; 41:1616–1626

488. Oberender F, Ganeshalingham A, Fortenberry JD, et al: Venoarterial extracorporeal membrane oxygenation versus conventional therapy in severe pediatric septic shock. Pediatr Crit Care Med 2018; 19:965–972

489. Ohlsson A, Lacy JB: Intravenous immunoglobulin for suspected or proven infection in neonates. Cochrane Database Syst Rev 2013; (7):CD001239

490. El-Nawawy A, El-Kinany H, Hamdy El-Sayed M, et al: Intravenous pol-yclonal immunoglobulin administration to sepsis syndrome patients: A prospective study in a pediatric intensive care unit. J Trop Pediatr 2005; 51:271–278

491. Brocklehurst P, Farrell B, King A, et al; INIS Collaborative Group: Treatment of neonatal sepsis with intravenous immune globulin. N Engl J Med 2011; 365:1201–1211

492. Alejandria MM, Lansang MA, Dans LF, et al: Intravenous immu-noglobulin for treating sepsis, severe sepsis and septic shock. Cochrane Database Syst Rev 2013; (9):CD001090

493. Berlot G, Vassallo MC, Busetto N, et al: Effects of the timing of ad-ministration of IgM- and IgA-enriched intravenous polyclonal immu-noglobulins on the outcome of septic shock patients. Ann Intensive Care 2018; 8:122

494. Parks T, Wilson C, Curtis N, et al: Polyspecific intravenous immu-noglobulin in clindamycin-treated patients with streptococcal toxic shock syndrome: A systematic review and meta-analysis. Clin Infect Dis 2018; 67:1434–1436

495. Kadri SS, Swihart BJ, Bonne SL, et al: Impact of intravenous im-munoglobulin on survival in necrotizing fasciitis with vasopressor-dependent shock: A propensity score-matched analysis from 130 US hospitals. Clin Infect Dis 2017; 64:877–885

496. Madsen MB, Lange T, Hjortrup PB, et al: Immunoglobulin for necro-tising soft tissue infections (INSTINCT): Protocol for a randomised trial. Dan Med J 2016; 63:A5250

497. Reveiz L, Guerrero-Lozano R, Camacho A, et al: Stress ulcer, gas-tritis, and gastrointestinal bleeding prophylaxis in critically ill pedi-atric patients: A systematic review. Pediatr Crit Care Med 2010; 11:124–132

498. Jimenez J, Drees M, Loveridge-Lenza B, et al: Exposure to gastric acid-suppression therapy is associated with health care- and com-munity-associated Clostridium difficile infection in children. J Pediatr Gastroenterol Nutr 2015; 61:208–211

499. Cook D, Heyland D, Griffith L, et al: Risk factors for clinically impor-tant upper gastrointestinal bleeding in patients requiring mechan-ical ventilation. Canadian Critical Care Trials Group. Crit Care Med 1999; 27:2812–2817

500. Duerksen DR: Stress-related mucosal disease in critically ill patients. Best Pract Res Clin Gastroenterol 2003; 17:327–344

501. Marik PE, Vasu T, Hirani A, et al: Stress ulcer prophylaxis in the new millennium: A systematic review and meta-analysis. Crit Care Med 2010; 38:2222–2228

502. Massicotte P, Julian JA, Gent M, et al; PROTEKT Study Group: An open-label randomized controlled trial of low molecular weight heparin for the prevention of central venous line-related thrombotic complications in children: The PROTEKT trial. Thromb Res 2003; 109:101–108

503. Brandao LR, Shah N, Shah PS: Low molecular weight heparin for prevention of central venous catheterization-related thrombosis in children. Cochrane Database Syst Rev 2014; (3):CD005982

504. van Ommen CH, Heijboer H, Büller HR, et al: Venous thromboembo-lism in childhood: A prospective two-year registry in The Netherlands. J Pediatr 2001; 139:676–681

505. Raffini L, Huang YS, Witmer C, et al: Dramatic increase in venous thromboembolism in children’s hospitals in the United States from 2001 to 2007. Pediatrics 2009; 124:1001–1008

506. Takemoto CM, Sohi S, Desai K, et al: Hospital-associated venous thromboembolism in children: Incidence and clinical characteristics. J Pediatr 2014; 164:332–338

Page 51: Surviving Sepsis Campaign International Guidelines for the ... · In 2001, the Surviving Sepsis Campaign (SSC) was formed by the Society of Critical Care Medicine (SCCM), European

Copyright © 2020 by the Society of Critical Care Medicine and the World Federation of Pediatric Intensive and Critical Care Societies.Unauthorized reproduction of this article is prohibited

Weiss et al

e102 www.pccmjournal.org February 2020 • Volume 21 • Number 2

Appendix 1. Summary of Guidelines

SCREENING, DIAGNOSIS, AND SYSTEMATIC MANAGEMENT OF SEPSIS

1. In children who present as acutely unwell, we suggest implementing systematic screening for timely recognition of septic shock and other sepsis-associated organ dysfunction (weak recommendation, very low quality of evidence). PICO 1 Remarks: Systematic screening needs to be tailored to the type of patients, resources, and procedures within each institution. Evaluation for the effectiveness and sustainability of screening should be incorporated as part of this process.

2. We were unable to issue a recommendation about using blood lactate values to stratify children with suspected septic shock or other sepsis-associated organ dysfunction into low-versus high-risk of having septic shock or sepsis. PICO 2

3. We recommend implementing a protocol/guideline for management of children with septic shock or other sepsis-associated organ dysfunction (BPS). PICO 3

4. We recommend obtaining blood cultures before initiating antimicrobial therapy in situations where this does not substantially delay antimicrobial administration (BPS). PICO 4

ANTIMICROBIAL THERAPY

5. In children with septic shock, we recommend starting antimicrobial therapy as soon as possible, within 1 hour of recognition (strong recommendation, very low quality of evidence). PICO 6

6. In children with sepsis-associated organ dysfunction but without shock, we suggest starting antimicrobial therapy as soon as possible after appropriate evaluation, within 3 hours of recognition (weak recommendation, very low quality of evidence). PICO 6

7. We recommend empiric broad-spectrum therapy with one or more antimicrobials to cover all likely pathogens (BPS). PICO 5

8.Once the pathogen(s) and sensitivities are available, we recommend narrowing empiric antimicrobial therapy coverage (BPS). PICO 5

9. If no pathogen is identified, we recommend narrowing or stopping empiric antimicrobial therapy according to clinical presentation, site of infection, host risk factors, and adequacy of clinical improvement in discussion with infectious disease and/or microbiological expert advice (BPS). PICO 5

10. In children without immune compromise and without high risk for multidrug-resistant pathogens, we suggest against the routine use of empiric multiple antimicrobials directed against the same pathogen for the purpose of synergy (weak recommendation, very low quality of evidence). PICO 8/9

Remarks: In certain situations, such as confirmed or strongly suspected group B streptococcal sepsis, use of empiric multiple antimicrobials directed against the same pathogen for the purpose of synergy may be indicated.

11. In children with immune compromise and/or at high risk for multidrug-resistant pathogens, we suggest using empiric multi-drug therapy when septic shock or other sepsis-associated organ dysfunction is present/suspected (weak recommendation, very low quality of evidence). PICO 8/9

12. We recommend using antimicrobial dosing strategies that have been optimized based on published pharmacokinetic/pharmacodynamic principles and with consideration of specific drug properties (BPS). PICO 7

13. In children with septic shock or sepsis-associated organ dysfunction who are receiving antimicrobials, we recommend daily assessment (e.g., clinical, laboratory assessment) for de-escalation of antimicrobial therapy (BPS). PICO 11

Remarks: This assessment should include a review of the ongoing indication for empiric antimicrobial therapy after the first 48 hours that is guided by microbiologic results and in response to clinical improvement and/or evidence of infection resolution. This recommendation applies to patients being treated with empiric, targeted, and combination therapy.

14. We recommend determining the duration of antimicrobial therapy according to the site of infection, microbial etiology, response to treatment, and ability to achieve source control (BPS). PICO 10

SOURCE CONTROL

15. We recommend that emergent source control intervention be implemented as soon possible after a diagnosis of an infection amenable to a source control procedure is made (BPS). PICO 12

Remarks: Appropriate diagnostic testing to identify the site of infection and microbial etiology should be performed, and advice from specialist teams (e.g., infectious diseases, surgery) should be sought, as appropriate, in order to prioritize interventions needed to achieve source control.

16. We recommend removal of intravascular access devices that are confirmed to be the source of sepsis or septic shock after other vascular access has been established and depending on the pathogen and the risks/benefits of a surgical procedure (strong recommendation, low quality of evidence). PICO 13

Page 52: Surviving Sepsis Campaign International Guidelines for the ... · In 2001, the Surviving Sepsis Campaign (SSC) was formed by the Society of Critical Care Medicine (SCCM), European

Copyright © 2020 by the Society of Critical Care Medicine and the World Federation of Pediatric Intensive and Critical Care Societies.Unauthorized reproduction of this article is prohibited

Online Special Article

Pediatric Critical Care Medicine www.pccmjournal.org e103

FLUID THERAPY

17. In healthcare systems with availability of intensive care, we suggest administering up to 40-60 mL/kg in bolus fluid (10-20 mL/kg per bolus) over the first hour, titrated to clinical markers of cardiac output and discontinued if signs of fluid overload develop, for the initial resuscitation of children with septic shock or other sepsis-associated organ dysfunction (weak recommendation, low quality of evidence). PICO 17

18. In healthcare systems with no availability of intensive care and in the absence of hypotension, we recommend against bolus fluid administration while starting maintenance fluids (strong recommendation, high quality of evidence). PICO 17

19. In healthcare systems with no availability of intensive care, if hypotension is present, we suggest administering up to 40 mL/kg in bolus fluid (10-20 mL/kg per bolus) over the first hour with titration to clinical markers of cardiac output and discontinued if signs of fluid overload develop (weak recommendation, low quality of evidence). PICO 17

Remarks: Clinical markers of cardiac output may include heart rate, blood pressure, capillary refill time, level of consciousness, and urine output. In all settings, the need for fluid administration should be guided by frequent reassessment of clinical markers of cardiac output, serial blood lactate measurement and advanced monitoring, when available. Signs of fluid overload that should limit further fluid bolus therapy may include clinical signs of pulmonary edema or new or worsening hepatomegaly.

20. We suggest using crystalloids, rather than albumin, for the initial resuscitation of children with septic shock or other sepsis-associated organ dysfunction (weak recommendation, moderate quality of evidence). PICO 15

Remarks: Although there is no difference in outcomes, this recommendation takes into consideration cost and other barriers of administering albumin compared to crystalloids.

21. We suggest using balanced/buffered crystalloids, rather than 0.9% saline, for the initial resuscitation of children with septic shock or other sepsis-associated organ dysfunction (weak recommendation, very low quality of evidence). PICO 14

22. We recommend against using starches in the acute resuscitation of children with septic shock or other sepsis-associated organ dysfunction (strong recommendation, moderate quality of evidence) PICO 16

23. We suggest against using gelatin in the resuscitation of children with septic shock or other sepsis-associated organ dysfunction (weak recommendation, low quality of evidence). PICO 16

HEMODYNAMIC MONITORING

24. We were unable to issue a recommendation about whether to target mean arterial blood pressure (MAP) at the 5th or 50th percentile for age in children with septic shock and other sepsis-associated organ dysfunction. PICO 21

25. We suggest not using bedside clinical signs in isolation to categorize septic shock in children as “warm” or “cold” (weak recommendation, very low quality of evidence). PICO 20

26. We suggest using advanced hemodynamic variables, when available, in addition to bedside clinical variables to guide the resuscitation of children with septic shock or other sepsis-associated organ dysfunction (weak recommendation, low quality of evidence). PICO 18

Remarks: Advanced hemodynamic monitoring may include cardiac output/cardiac index, systemic vascular resistance, or central venous oxygen saturation (ScvO2).

27. We suggest using trends in blood lactate levels, in addition to clinical assessment, to guide resuscitation of children with septic shock and other sepsis-associated organ dysfunction (weak recommendation, very low quality of evidence). PICO 19

Remarks: In children with an elevated blood lactate, repeat testing that reveals a persistent elevation in blood lactate may indicate incomplete hemodynamic resuscitation and should prompt efforts, as needed, to further promote hemodynamic stability

VASOACTIVE MEDICATIONS

28. We suggest using epinephrine, rather than dopamine, in children with septic shock (weak recommendation, low quality of evidence). PICO 22

29.We suggest using norepinephrine, rather than dopamine, in children with septic shock (weak recommendation, very low quality of evidence). PICO 23

30. We were unable to issue a recommendation for a specific first-line vasoactive infusion for children with septic shock. PICO 22/23

31. We were unable to issue a recommendation about initiating vasoactive agents through peripheral access in children with septic shock. PICO 26

Remarks: It is reasonable to begin vasoactive infusions after 40-60 mL/kg of fluid resuscitation if the patient continues to have evidence of abnormal perfusion. Either epinephrine or norepinephrine may be administered through a peripheral vein (or intraosseous, if in place) if central venous access is not readily accessible. Dopamine may be substituted as the first-line vasoactive infusion, administered either peripherally or centrally, if epinephrine or norepinephrine is not readily available.

Page 53: Surviving Sepsis Campaign International Guidelines for the ... · In 2001, the Surviving Sepsis Campaign (SSC) was formed by the Society of Critical Care Medicine (SCCM), European

Copyright © 2020 by the Society of Critical Care Medicine and the World Federation of Pediatric Intensive and Critical Care Societies.Unauthorized reproduction of this article is prohibited

Weiss et al

e104 www.pccmjournal.org February 2020 • Volume 21 • Number 2

32. We suggest either adding vasopressin or further titrating catecholamines in children with septic shock who require high-dose catecholamines (weak recommendation, low quality of evidence). PICO 25

Remarks: No consensus was achieved on the optimal threshold for initiating vasopressin. Therefore, this decision should be made according to individual clinician preference.

33. We were unable to issue a recommendation about adding an inodilator in children with septic shock and cardiac dysfunction despite other vasoactive agents. PICO 24

VENTILATION

34. We were unable to issue a recommendation about whether to intubate children with fluid-refractory, catecholamine-resistant septic shock. PICO 27

35.We suggest not to use etomidate when intubating children with septic shock or other sepsis-associated organ dysfunction (weak recommendation, low quality of evidence). PICO 28

36. We suggest a trial of non-invasive mechanical ventilation (over invasive mechanical ventilation) in children with sepsis-induced pediatric ARDS (PARDS) without a clear indication for intubation and who are responding to initial resuscitation (weak recommendation, very low quality of evidence). PICO 29 Remarks: When non-invasive mechanical ventilation is initiated, clinicians should carefully and frequently re-evaluate the patient’s condition.

37. We suggest using high positive end-expiratory pressure (PEEP) in children with sepsis-induced PARDS (weak recommendation, very low quality of evidence). PICO 30

Remarks: The exact level of high PEEP has not been tested or determined in PARDS patients. Some RCTs and observational studies in PARDS have used and advocated for use of the ARDS-network PEEP to fractional inspired oxygen (FiO2) grid though adverse hemodynamic effects of high PEEP may be more prominent in children with septic shock.

38. We cannot suggest for or against the use of recruitment maneuvers in children with sepsis-induced PARDS and refractory hypoxemia. PICO 31 Remarks: If a recruitment maneuver is considered, the use of a stepwise, incremental and decremental PEEP titration maneuver is preferred over sustained inflation techniques that have not been optimized through direct testing in PARDS patients. All PARDS patients must be carefully monitored for tolerance of the maneuver.

39. We suggest a trial of prone positioning in children with sepsis and severe PARDS (weak recommendation, low quality of evidence). PICO 32

Remarks: Research trials in adults with ARDS and children with PARDS have emphasized prone positioning for at least 12 hours per day, as tolerated.

40. We recommend against the routine use of inhaled nitric oxide (iNO) in all children with sepsis-induced PARDS (strong recommendation, low quality of evidence). PICO 33

41. We suggest using iNO as a rescue therapy in children with sepsis-induced PARDS and refractory hypoxemia after other oxygenation strategies have been optimized (weak recommendation, moderate quality of evidence). PICO 33

42. We were unable to issue a recommendation to use high-frequency oscillatory ventilation (HFOV) versus conventional ventilation in children with sepsis-induced PARDS. PICO 34

43. We suggest using neuromuscular blockade in children with sepsis and severe PARDS (weak recommendation, very low quality of evidence). PICO 35

Remarks: The exact duration of neuromuscular blockade use in severe PARDS patients has not been determined to date. Most of the adult RCT data and pediatric observational data support treatment for 24-48 hours after ARDS onset.

CORTICOSTEROIDS

44. We suggest against using intravenous hydrocortisone to treat children with septic shock if adequate fluid resuscitation and vasopressor therapy are able to restore hemodynamic stability (weak recommendation, low quality of evidence). PICO 47

45. We suggest that either intravenous hydrocortisone or no hydrocortisone may be used if adequate fluid resuscitation and vasopressor therapy are not able to restore hemodynamic stability (weak recommendation, low quality of evidence). PICO 47

ENDOCRINE AND METABOLIC

46. We recommend against insulin therapy to maintain glucose target at or below 140 mg/dL (7.8 mmol/L) (strong recommendation, moderate quality of evidence). PICO 52/60

47. We were unable to issue a recommendation regarding what blood glucose range to target for children with septic shock and other sepsis-associated organ dysfunction. PICO 52/60

Page 54: Surviving Sepsis Campaign International Guidelines for the ... · In 2001, the Surviving Sepsis Campaign (SSC) was formed by the Society of Critical Care Medicine (SCCM), European

Copyright © 2020 by the Society of Critical Care Medicine and the World Federation of Pediatric Intensive and Critical Care Societies.Unauthorized reproduction of this article is prohibited

Online Special Article

Pediatric Critical Care Medicine www.pccmjournal.org e105

48. We were unable to issue a recommendation as to whether to target normal blood calcium levels in children with septic shock or sepsis-associated organ dysfunction. PICO 62

49. We suggest against the routine use of levothyroxine in children with septic shock and other sepsis-associated organ dysfunction in a sick euthyroid state (weak recommendation, low quality of evidence). PICO 63

50. We suggest either antipyretic therapy or a permissive approach to fever in children with septic shock or other sepsis-associated organ dysfunction (weak recommendation, moderate quality of evidence). PICO 64

NUTRITION

51. We were unable to issue a recommendation regarding early hypocaloric/trophic enteral feeding followed by slow increase to full enteral feeding versus early full enteral feeding in children with septic shock or sepsis-associated organ dysfunction without contraindications to enteral feeding. PICO 51

52. We suggest not withholding enteral feeding solely on the basis of vasoactive-inotropic medication administration (weak recommendation, low quality of evidence). PICO 48 Remarks: Enteral feeding is not contraindicated in children with septic shock after adequate hemodynamic resuscitation who no longer require escalating doses of vasoactive agents or in whom weaning of vasoactive agents has started.

53. We suggest enteral nutrition as the preferred method of feeding and that parenteral nutrition may be withheld in the first 7 days of PICU admission in children with septic shock or other sepsis-associated organ dysfunction (weak recommendation, moderate quality of evidence). PICO 49/50

54. We suggest against supplementation with specialized lipid emulsions in children with septic shock or other sepsis-associated organ dysfunction (weak recommendation, very low quality of evidence). PICO 53

55. We suggest against the routine measurements of gastric residual volumes (GRV) in children with septic shock or other sepsis-associated organ dysfunction (weak recommendation, low quality of evidence). PICO 54

56. We suggest administering enteral feeds through a gastric tube, rather than a post-pyloric feeding tube, to children with septic shock or other sepsis-associated organ dysfunction who have no contraindications to enteral feeding (weak recommendation, low quality of evidence). PICO 55

57. We suggest against the routine use of prokinetic agents for the treatment of feeding intolerance in children with septic shock or other sepsis-associated organ dysfunction (weak recommendation, low quality of evidence). PICO 56

58. We suggest against the use of selenium in children with septic shock or other sepsis-associated organ dysfunction (weak recommendation, low quality of evidence). PICO 57

59. We suggest against the use of glutamine supplementation in children with septic shock or other sepsis-associated organ dysfunction (weak recommendation, low quality of evidence). PICO 58

60. We suggest against the use of arginine in the treatment of children with septic shock or other sepsis-associated organ dysfunction (weak recommendation, very low quality of evidence). PICO 59

61. We suggest against using zinc supplementation in children with septic shock and other sepsis-associated organ dysfunction (weak recommendation, very low quality of evidence). PICO 61

62. We suggest against the use of ascorbic acid (vitamin C) in the treatment of children with septic shock or other sepsis-associated organ dysfunction (weak recommendation, very low quality of evidence). PICO 65

63. We suggest against the use of thiamine to treat children with sepsis-associated organ dysfunction (weak recommendation, low quality of evidence). PICO 66

64. We suggest against the acute repletion of vitamin D deficiency (VDD) for treatment of septic shock or other sepsis-associated organ dysfunction (weak recommendation, very low quality of evidence). PICO 67

BLOOD PRODUCTS

65. We suggest against transfusion of red blood cells if the blood hemoglobin concentration is ≥7 g/dL in hemodynamically stabilized children with septic shock or other sepsis-associated organ dysfunction (weak recommendation, low quality of evidence). PICO 38 Remarks: According to the 2018 Transfusion and Anemia Expertise Initiative (TAXI) guidelines, for the purposes of red blood cell transfusion, “hemodynamically stabilized” is defined as a mean arterial blood pressure higher than 2 standard deviations below normal for age and no increase in vasoactive medications for at least 2 hours.

66. We cannot make a recommendation regarding hemoglobin transfusion thresholds for critically ill children with unstable septic shock. PICO 38

67. We suggest against prophylactic platelet transfusion based solely on platelet levels in non-bleeding children with septic shock or other sepsis-associated organ dysfunction and thrombocytopenia (weak recommendation, very low quality of evidence). PICO 40

Page 55: Surviving Sepsis Campaign International Guidelines for the ... · In 2001, the Surviving Sepsis Campaign (SSC) was formed by the Society of Critical Care Medicine (SCCM), European

Copyright © 2020 by the Society of Critical Care Medicine and the World Federation of Pediatric Intensive and Critical Care Societies.Unauthorized reproduction of this article is prohibited

Weiss et al

e106 www.pccmjournal.org February 2020 • Volume 21 • Number 2

68. We suggest against prophylactic plasma transfusion in non-bleeding children with septic shock or other sepsis-associated organ dysfunction and coagulation abnormalities (weak recommendation, very low quality of evidence). PICO 39 Remarks: Prophylactic plasma transfusion refers to situations in which there is an abnormality in laboratory coagulation testing but no active bleeding.

PLASMA EXCHANGE, RENAL REPLACEMENT, AND EXTRACORPOREAL SUPPORT

69. We suggest against using plasma exchange in children with septic shock or other sepsis-associated organ dysfunction without thrombocytopenia-associated multiple organ failure (TAMOF) (weak recommendation, very low quality of evidence). PICO 37

70. We cannot suggest for or against the use of plasma exchange in children with septic shock or other-sepsis-associated organ dysfunction with TAMOF. PICO 37

71. We suggest using renal replacement therapy to prevent or treat fluid overload in children with septic shock or other sepsis-associated organ dysfunction who are unresponsive to fluid restriction and diuretic therapy (weak recommendation, very low quality of evidence). PICO 43

72. We suggest against high-volume hemofiltration over standard hemofiltration in children with septic shock or other sepsis-associated organ dysfunction who are treated with renal replacement therapy (weak recommendation, low quality of evidence). PICO 44

73. We suggest using veno-venous (VV) extracorporeal membrane oxygenation (ECMO) in children with sepsis-induced PARDS and refractory hypoxia (weak recommendation, very low quality of evidence). PICO 36

74. We suggest using veno-arterial (VA) ECMO as a rescue therapy in children with septic shock only if refractory to all other treatments (weak recommendation, very low quality of evidence). PICO 45

IMMUNOGLOBULINS

75. We suggest against the routine use of intravenous immune globulin (IVIG) in children with septic shock or other sepsis-associated organ dysfunction (weak recommendation, low quality of evidence). PICO 46 Remarks: Although routine use of IVIG is not recommended, select patients may benefit from such treatment.

PROPHYLAXIS

76. We suggest against the routine use of stress ulcer prophylaxis in critically ill children with septic shock or other sepsis-associated organ dysfunction, except for high-risk patients (weak recommendation, very low quality of evidence). PICO 41

Remarks: Although routine stress-ulcer prophylaxis is not recommended, some high-risk patients may benefit from stress ulcer prophylaxis. Studies have supported benefit of stress ulcer prophylaxis when baseline rate of clinically important bleeding is approximately 13%.

77. We suggest against routine deep vein thrombosis (DVT) prophylaxis (mechanical or pharmacologic) in critically ill children with septic shock or other sepsis-associated organ dysfunction, but potential benefits may outweigh risks and costs in specific populations (weak recommendation, low quality of evidence). PICO 42